WO2025137751A1 - Polynucleotide, chimeric antigen receptor, vector, composition, method for producing a modified immune effector cell and use of the polynucleotide - Google Patents
Polynucleotide, chimeric antigen receptor, vector, composition, method for producing a modified immune effector cell and use of the polynucleotide Download PDFInfo
- Publication number
- WO2025137751A1 WO2025137751A1 PCT/BR2024/050577 BR2024050577W WO2025137751A1 WO 2025137751 A1 WO2025137751 A1 WO 2025137751A1 BR 2024050577 W BR2024050577 W BR 2024050577W WO 2025137751 A1 WO2025137751 A1 WO 2025137751A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cells
- domain
- polynucleotide
- car
- seq
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
Definitions
- POLYNUCLEOTIDE CHIMERIC ANTIGEN RECEPTOR, VECTOR, COMPOSITION, METHOD FOR PRODUCTION OF MODIFIED EFFECTOR IMMUNE CELL AND USE OF POLYNUCLEOTIDE
- the present invention relates to a polynucleotide encoding an anti-BCMA chimeric antigen receptor (CAR) and the polypeptide corresponding to the anti-BCMA chimeric antigen receptor (CAR) itself.
- the present invention also relates to a vector and a composition comprising an effector immune cell comprising the polynucleotide, in addition to a method for producing a modified effector immune cell and the use of the polynucleotide, the vector, the composition or the effector immune cell produced by the method for the manufacture of a medicament for the treatment of multiple myeloma.
- MM Multiple myeloma
- BM bone marrow
- MGUS monoclonal gammopathy of undetermined significance
- M protein monoclonal protein
- the diagnosis of multiple myeloma is defined when one or more symptoms are observed, including hypercalcemia, renal failure, anemia, elevated serum M protein and lytic bone lesions, and 10% or more plasma cells in the bone marrow or plasmacytoma identified by biopsy (Rajkumar et al., 2014; Rajkumar, SV, 2022).
- Treatment options aim to reduce the abundance of malignant plasma cells in BM and include procedures such as hematopoietic stem cell transplantation (HSCT), radiotherapy, and chemotherapy.
- HSCT hematopoietic stem cell transplantation
- radiotherapy radiotherapy
- chemotherapy chemotherapy
- new treatment options such as 2nd generation proteasome inhibitors, 2nd generation immunomodulatory drugs, and anti-CD38 monoclonal antibodies, have resulted in a significant improvement in the survival of patients with multiple myeloma (Cowan et al., 2022; Rodr ⁇ guez-Otero et al., 2020).
- the disease has a relapsing and progressive profile, leading to refractoriness or relapse in most cases.
- the main objective of treating multiple myeloma is to prolong patient survival and the time until disease progression.
- Treatment options for relapsed or refractory MM include anti-CD38 or anti-CD319 monoclonal antibodies, steroids, immunomodulatory drugs, proteasome inhibitors, alkylating agents, antimetabolic agents, and transplantation (Cowan et al., 2022; Shah et al., 2012).
- the treatment of multiple myeloma generates high costs for health systems and operators in the country and worldwide, as the management of the disease is expensive.
- immunotherapy such as the use of CAR-T and/or CAR-NK cells, can provide great help in several aspects (K ⁇ hler et al., 2018; Perez-Amill et al., 2021).
- a CAR receptor chimeric antigen receptor
- a CAR receptor consists of three linked domains: an extracellular recognition domain, a transmembrane domain, and intracellular signaling domains (also known as the cytoplasmic domain).
- the extracellular domain is formed by variable portions of the light and heavy chains of an immunoglobulin specific for the recognition of an antigen of choice, optionally linked by a flexible linker (Ramos, C.A., Dotti, G., 2011).
- This extracellular portion is linked to a transmembrane domain, which in turn binds to the intracellular signaling domain.
- the link between the extracellular domain and the transmembrane domain is called the hinge (Miliotou, AN, Papadopoulou, LC, 2018).
- the intracellular domain can combine signaling domains of the TCR receptor complex and costimulatory molecules of T lymphocytes. These costimulatory molecules are necessary to increase the proliferation, cytotoxicity and persistence of these cells in vitro and in vivo.
- CAR cell therapy combines the high specificity of monoclonal antibodies with the potent cytotoxicity and long-term persistence of cytotoxic T cells (Barrett et al., 2014; Zhang et al., 2017).
- CAR-T cell therapy is a type of treatment in which the patient's T cells are genetically modified so that they become specific and effective in attacking cancer cells.
- T lymphocytes are usually collected from the patient and a gene encoding a CAR is inserted into them. This receptor specifically recognizes a protein present in the patient's tumor cells, increasing the affinity of the T lymphocytes (in this case, called CAR-T) for the tumor cells.
- CAR-T T lymphocytes
- These cells are then expanded and infused into the patient, where they continue to expand and can recognize and kill cancer cells (Picanto-Castro et al., 2020; Miliotou et al., 2018).
- NK cells from the patient or another donor are genetically modified to attack cancer cells. Similar to T cells, a gene encoding a CAR is inserted into the NK cells, which are then expanded and infused into the patient, where they continue to expand and can recognize and kill cancer cells.
- CAR-NK cell therapy emerges as an alternative to CAR-T cells to overcome some limitations such as (i) the difficulty in preparing clinically relevant doses of autologous CAR-T cells in pretreated lymphopenic patients or (ii) the risk of developing graft-versus-host disease (GVHD) associated with the use of T cells collected from an allogeneic source, that is, from another donor (Mehta et al., 2018; Goulmy, E., 1997).
- GVHD graft-versus-host disease
- NK cells represent the most effective effectors against tumors with a mechanism of action distinct from that of T cells (Davies et al., 2014; Vivier et al., 2012).
- NK cells In contrast to other lymphocytes, NK cells do not express antigen-specific receptors, but rather inactive germline-encoded receptors that are either activators or inhibitors that can induce either a positive or negative signal, and the balance of these signals controls the effector function of NK cells (Lanier, LL, 1998). Mature NK cells also have a relatively limited life span, allowing effective antitumor activity but simultaneously reducing the likelihood of long-term adverse events, such as prolonged cytopenias, due to recognition in non-tumor targets (normal tissues), such as B-cell aplasia (Maude et al., 2014). Furthermore, CAR-NK cells maintain their intrinsic ability to recognize tumor cells through their native receptors.
- tumor cells are less likely to escape the immune surveillance of NK cells, even if tumor cells downregulate the CAR target antigen (Mehta et al., 2018; Sotillo et al., 2015).
- HSCT hematopoietic stem cell transplantation
- the first generation consists of the extracellular domain fused to a CD3 zeta cytoplasmic domain. Although they are effective in vitro, this has not been observed in in vivo assays.
- Second-generation CARs have an additional costimulatory signaling domain, while third-generation CARs have two or more of these costimulatory domains.
- the addition of costimulatory domains generates greater proliferation, cytotoxicity, persistence and clinical efficacy (Ramos, CA, Dotti, G., 2011 ; Essand, M., Loskog, ASI, 2013).
- 3rd generation CARs aims to further increase the potency of CAR-T lymphocytes, but trials comparing 2nd and 3rd generation CARs have shown controversial results, so that the superiority of 3rd generation CARs over 2nd generation CARs has not yet been proven.
- 4th generation CARs present the components previously listed with the addition of other genes, such as cytokines, with the aim of increasing the activity of the cells in fighting the tumor.
- cytokines genes that are associated with the aim of increasing the activity of the cells in fighting the tumor.
- 3 the domain of the [3 chain of the cytoplasmic portion of the IL-2 receptor (IL-2R
- target antigen for CAR cell therapy is an extremely important step for successful treatment.
- the ideal target antigen should be strongly expressed in the target cells of the treatment, so that the CAR cells can easily locate the tumor cells in order to eliminate them.
- the antigen should be exclusively expressed in the target cells, or have low expression in other cell types. This is important so that the CAR cells do not exert their function in healthy or nonspecific tissues, avoiding adverse effects (Wei et al., 2019).
- the objective of the present invention is to provide polynucleotides encoding anti-BCMA CARs with cytotoxic activity for multiple myeloma cells, as well as a vector and an effector immune cell comprising the same, which can be used for the development of more efficient CAR-T and/or CAR-NK cell immunotherapies, seeking to alleviate the operational difficulties and/or adverse effects of the product.
- the inventors have surprisingly managed to overcome the problems of the prior art by means of polynucleotides encoding anti-BCMA chimeric antigen receptors (CARs), in which the CARs comprise an antigen-binding domain, a transmembrane domain and an intracellular domain comprising one or more costimulatory domains and one or more signaling domains, in which the anti-BCMA binding domain comprises the light chain variable domain of SEQ ID NO: 1 and the heavy chain variable domain of SEQ ID NO: 3.
- CARs anti-BCMA chimeric antigen receptors
- the inventors have also developed a vector and a composition, comprising an effector immune cell, comprising the polynucleotide, in addition to a method for the production of a modified effector immune cell and the use of the polynucleotide, the vector, the composition or the effector immune cell produced by the method for the manufacture of a medicament for the treatment of multiple myeloma.
- the present invention relates to a polynucleotide encoding an anti-BCMA chimeric antigen receptor (CAR) and to a polypeptide corresponding to the anti-BCMA chimeric antigen receptor (CAR) itself.
- the present invention also relates to a vector and a composition, comprising an effector immune cell, which comprise the polynucleotide.
- the present invention is also directed to a method for producing a modified effector immune cell and to the use of the polynucleotide, vector, composition or effector immune cell produced by the method for manufacturing a medicament for the treatment of multiple myeloma.
- Figures 1 a, 1 b and 1c show the anti-BCMA CAR expression/transfer vectors of the present invention.
- Figure 2 shows the packaging cassette plasmid (psPAX2 vector) of the vector of the present invention.
- Figure 3 shows the envelope cassette plasmid (pMD2.G vector) of the vector of the present invention.
- Figure 4 demonstrates the gating strategy for T lymphocyte immunophenotyping.
- Figure 5 shows the results of pre- and post-selection T lymphocyte immunophenotyping with magnetic beads.
- Figure 6 is a representative analysis of T lymphocyte activation after stimulation with Dynabeads CD3/CD28 for 24 hours.
- Figure 7 demonstrates the gating strategy for CAR+ lymphocyte detection with the BCMA CAR Detection kit.
- Figure 8 shows the transduction efficiency of anti-BCMA, anti-BCMA(IL-15), and anti-CD19 CARs.
- Figure 9 depicts the viability and expansion of CAR-T lymphocytes in culture for 15 days.
- Figure 10 corresponds to the experimental design of the calcein assay.
- Figure 11 corresponds to the cell lysis curves obtained in the calcein assays of CAR-T cells and non-transduced lymphocytes co-cultured with two tumor cell lines, A) MM1 S (BCMA+ CD19-) and B) Nalm6 (BCMA-CD19+).
- Figure 12 demonstrates the gating strategy for cytokine production assay analysis.
- Figure 13 shows the production of the cytokines IFN- ⁇ (A) and TNF- ⁇ (B) by populations of anti-BCMA CAR-T lymphocytes (with and without IL-15), non-transduced lymphocytes and CD19 CAR-T lymphocytes in response to stimulation by PMA + lonomycin, or when co-cultured with the MM1 S and Nalm6 tumor lines.
- Figure 14 represents the gating strategy used for immunophenotyping of NK cells pre- and post-selection.
- Figure 15 exemplifies the gating strategy used to analyze NK cells expressing the anti-BCMA-IL-15 CAR.
- Figure 16 depicts the transduction efficiency of anti-BCMA-IL-15 CAR for NK and memory-like (ML) NK cells.
- Figure 17 shows the transduction efficiency of anti-BCMA-IL-15 CAR for NK and memory-like NK cells.
- Figure 20 shows the production of IFN-y, TNF-a and CD107a by anti-BCMA-IL-15 NK and NK memory like CAR cells in response to stimulation by PMA + lonomycin, or in co-culture with the MM1 S tumor cell line.
- the light and heavy chain variable domains of SEQ ID Nos: 1 and 3 correspond to the light and heavy chain variable domains of the rabbit monoclonal anti-murine BCMA antibody, clone 11 D5.3.
- the light and heavy chain variable domains are encoded by the nucleotide sequences of SEQ ID Nos: 2 and 4, respectively, and degenerate sequences thereof.
- the encoded CAR anti-BCMA binding domain is a Single-Chain Fragment Variable (ScFv), wherein the light and heavy chain variable domains can be in either of the following orientations: light chain variable domain-linker-heavy chain variable domain or heavy chain variable domain-linker-light chain variable domain.
- the orientation of the ScFv is light chain variable domain-linker-heavy chain variable domain.
- the orientation of the ScFv is heavy chain variable domain-linker-light chain variable domain.
- the ScFv comprises the sequence of SEQ ID NO: 9, encoded by SEQ ID NO: 10. ID NO: 10 and its degenerate sequences.
- the encoded anti-BCMA CAR includes a transmembrane domain comprising a transmembrane domain of a protein, e.g., selected from the group consisting of the T-cell receptor alpha, beta, or zeta chain, CD28, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, and CD 154.
- the encoded transmembrane domain comprises the CD8 transmembrane domain.
- the encoded transmembrane domain comprises the CD8 alpha transmembrane domain, comprising the sequence of SEQ ID NO: 11, encoded by SEQ ID NO: 12, and degenerate sequences thereof.
- the encoded anti-BCMA binding domain is connected to the transmembrane domain by a hinge region.
- the hinge region comprises the constant region of an IgG1 molecule, a CD8 or a CD28 molecule.
- the hinge region comprises CD8 alpha.
- the hinge region comprises the sequence of SEQ ID NO: 13, encoded by SEQ ID NO: 14 and degenerate sequences thereof.
- the encoded anti-BCMA CAR includes a signal peptide.
- the signal peptide comprises an lgG1 heavy chain signal peptide, a granulocyte-macrophage colony-stimulating factor receptor 2 (GM-CSFR2) signal peptide, or a CD8 alpha signal peptide.
- the signal peptide comprises a CD8 alpha signal peptide.
- the signal peptide comprises the sequence of SEQ ID NO: 15, encoded by SEQ ID NO: 16, and degenerate sequences thereof.
- the encoded anti-BCMA CAR includes one or more costimulatory domains to increase cell efficacy and expansion. expressing CAR receptors.
- costimulatory domain refers to an intracellular signaling domain of a costimulatory molecule. Costimulatory molecules are cell surface molecules other than antigen receptors or Fc receptors that provide a second signal necessary for NK/T cell activation and function following antigen binding.
- the one or more costimulatory domains are selected from CD28, CD27, OX-40 (CD134), DAP10, DAP 12, 4-1 BB (CD137), CD40L, 2B4, DNAM, CS1, CD48, NKG2D, NKp30, NKp44, NKp46, NKp80, and a combination thereof.
- the costimulatory domain comprises 4-1 BB.
- the costimulatory domain comprises the sequence of SEQ ID NO: 17, encoded by SEQ ID NO: 18 and by degenerate sequences thereof.
- the costimulatory domain comprises 2B4.
- the costimulatory domain comprises the sequence of SEQ ID NO: 19, encoded by SEQ ID NO: 20 and by degenerate sequences thereof.
- the encoded anti-BCMA CAR includes an intracellular signaling domain that regulates primary activation of the TCR complex in a stimulatory or inhibitory manner.
- the encoded anti-BCMA CAR comprises a CD3 zeta primary signaling domain.
- the CD3 zeta primary signaling domain comprises the sequence of SEQ ID NO: 21, encoded by SEQ ID NO: 22 and degenerate sequences thereof.
- the encoded anti-BCMA CAR includes a CD3 zeta primary signaling domain and one or more costimulatory signaling domains, which may be linked in any order to the C-terminus of the transmembrane domain.
- the encoded anti-BCMA CAR includes one or more interleukins.
- the encoded anti-BCMA CAR includes IL-2, IL-7, IL-15 or a combination thereof.
- IL-2 is the main T-cell stimulating factor, as a growth and activation factor for all T lymphocyte subpopulations.
- IL-7 is a cytokine that stimulates the growth and maturation of B lymphocytes and the activation of T lymphocytes, and is secreted by bone marrow and thymus stromal cells.
- IL-15 is a cytokine with structural similarity to IL-2 that binds via a complex composed of IL-2/IL-15 receptor beta chain and common gamma chain. IL-15 is secreted by mononuclear phagocytes after virus infection.
- the encoded anti-BCMA CAR includes IL-15.
- the IL-15 cytokine comprises the sequence of SEQ ID NO: 23, encoded by SEQ ID NO: 24 and degenerate sequences thereof.
- the encoded anti-BCMA CAR includes IL-2.
- the IL-2 cytokine comprises the sequence of SEQ ID NO: 25, encoded by SEQ ID NO: 26 and degenerate sequences thereof.
- the encoded anti-BCMA CAR signaling domain is connected to interleukin via a cleavage sequence.
- the cleavage sequence comprises 2A sequence elements or 2S-like sequence elements, which can be used for protein binding or co-expression. It is common knowledge in the art that cleavage sequences can be used to co-express genes by linking open reading frames to form a single cistron.
- the cleavage sequence comprised in the encoded anti-BCMA CAR of the present invention comprises equine rhinitis virus (E2A), foot-and-mouth disease virus (F2A), Thosea asigna virus (T2A), porcine Teschovirus-1 (P2A), or a combination thereof.
- the cleavage sequence comprises T2A.
- the T2A cleavage sequence comprises the sequence of SEQ ID NO: 27, encoded by SEQ ID NO: 28, and degenerate sequences thereof.
- the cleavage sequence comprises P2A.
- the P2A cleavage sequence comprises the sequence of SEQ ID NO: 29, encoded by SEQ ID NO: 30, and degenerate sequences thereof.
- the cleavage sequence comprises E2A.
- the E2A cleavage sequence comprises the sequence of SEQ ID NO: 31, encoded by SEQ ID NO: 32 and degenerate sequences thereof.
- the encoded anti-BCMA CAR comprises a CD8 signal peptide, a linker, a CD8 hinge region, a CD8 transmembrane domain, a 4-1 BB costimulatory domain, and a CD3 zeta signaling domain.
- the encoded anti-BCMA CAR comprises a CD8 signal peptide, a linker, a CD8 hinge region, a CD8 transmembrane domain, a 4-1 BB costimulatory domain, a CD3 zeta signaling domain, a T2A cleavage sequence, and IL-15.
- the encoded anti-BCMA CAR comprises a CD8 signal peptide, a linker, a CD8 hinge region, a CD8 transmembrane domain, a 2B4 costimulatory domain, a CD3 zeta signaling domain, a T2A cleavage sequence, and IL-15.
- the encoded anti-BCMA CAR comprises a combination of the sequences selected from the odd sequences of SEQ ID NOs: 1 to 31.
- the encoded anti-BCMA CAR comprises one of SEQ ID NOs: 33, 35 or 37. In a more preferred embodiment, the encoded anti-BCMA CAR comprises SEQ ID NO: 33. In a more preferred embodiment, the encoded anti-BCMA CAR comprises SEQ ID NO: 35. In a more preferred embodiment, the encoded anti-BCMA CAR comprises SEQ ID NO: 37.
- the polynucleotide encoding the anti-BCMA CAR of the present invention comprises a combination of the nucleotide sequences selected from the even sequences of SEQ ID NOs: 2 to 32.
- the polynucleotide encoding the anti-BCMA CAR of the present invention comprises one of SEQ ID NOs: 34, 36 or 38. In a more preferred embodiment, the polynucleotide encoding the anti-BCMA CAR of the present invention comprises SEQ ID NO: 34. In another more preferred embodiment, the polynucleotide encoding the anti-BCMA CAR of the present invention comprises SEQ ID NO: 36. In another more preferred embodiment, the polynucleotide encoding the anti-BCMA CAR of the present invention comprises SEQ ID NO: 38.
- the present invention also relates to a polypeptide corresponding to the chimeric antigen receptor (CAR) comprising an anti-BCMA binding domain, a transmembrane domain and an intracellular domain comprising one or more costimulatory domains and a signaling domain, wherein the anti-BCMA binding domain comprises the ⁇ -chain variable domain.
- CAR chimeric antigen receptor
- the light and heavy chain variable domains of SEQ ID NOs: 1 and 3 of the anti-BCMA CAR of the present invention correspond to the light and heavy chain variable domains of the rabbit monoclonal anti-murine BCMA antibody, clone 11 D5.3.
- the BCMA-binding variable domains of the CAR of the present invention are connected by a flexible peptide linker.
- the peptide linker is selected from GSTSGSGKPGSGEGSTKG, (G4S)3, (G4S)4, among others.
- the peptide linker is GSTSGSGKPGSGEGSTKG (SEQ ID NO: 5).
- the peptide linker is (G4S)3 (SEQ ID NO: 7).
- the anti-BCMA binding domain of the CAR of the present invention is a ScFv, wherein the light and heavy chain variable domains can be in any of the following orientations: light chain variable domain-linker-heavy chain variable domain or heavy chain variable domain-linker-light chain variable domain.
- the orientation of the ScFv is light chain variable domain-linker-heavy chain variable domain.
- the orientation of the ScFv is heavy chain variable domain-linker-light chain variable domain.
- the ScFv comprises the sequence of SEQ ID NO: 9.
- the anti-BCMA CAR of the present invention includes a transmembrane domain comprising a transmembrane domain of a protein, e.g., selected from the alpha, beta or zeta chain of the T cell receptor, CD28, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137 and CD 154.
- the anti-BCMA CAR of the present invention includes a signal peptide.
- the signal peptide comprises an lgG1 heavy chain signal peptide, granulocyte-macrophage colony-stimulating factor receptor 2 (GM-CSFR2) signal peptide, or a CD8 alpha signal peptide.
- the signal peptide comprises a CD8 alpha signal peptide.
- the signal peptide of the anti-BCMA CAR of the present invention comprises the sequence of SEQ ID NO: 15.
- the anti-BCMA CAR of the present invention includes one or more costimulatory domains to increase the efficacy and expansion of cells expressing the same.
- the one or more costimulatory domains of the anti-BCMA CAR of the present invention are selected from, but not limited to, CD28, CD27, OX-40 (CD134), DAP10, DAP 12, 4-1 BB (CD137), CD40L, 2B4, DNAM, CS1, CD48, NKG2D, NKp30, NKp44, NKp46, NKp80, and a combination thereof.
- the costimulatory domain of the anti-BCMA CAR of the present invention comprises 4-1 BB.
- the 4-1 BB costimulatory domain comprises sequence of SEQ ID NO: 17.
- the costimulatory domain of the anti-BCMA CAR of the present invention comprises 2B4.
- the costimulatory domain 2B4 comprises the sequence of SEQ ID NO: 19.
- the anti-BCMA CAR of the present invention includes an intracellular signaling domain.
- the anti-BCMA CAR of the present invention comprises a CD3 zeta primary signaling domain.
- the CD3 zeta primary signaling domain comprises the sequence of SEQ ID NO: 21.
- the anti-BCMA CAR of the present invention includes a CD3 zeta primary signaling domain and one or more costimulatory signaling domains, which may be linked in any order to the C-terminus of the transmembrane domain.
- the anti-BCMA CAR of the present invention further includes a cytokine.
- the anti-BCMA CAR of the present invention includes interleukin, interferon- ⁇ (IFN- ⁇ ), or a combination thereof.
- the anti-BCMA CAR of the present invention includes one or more interleukins.
- the CAR of the present invention includes IL-2, IL-7, IL-15 or a combination thereof.
- the anti-BCMA CAR of the present invention includes IL-15.
- the IL-15 cytokine comprises the sequence of SEQ ID NO: 23.
- the anti-BCMA CAR includes IL-2.
- the IL-2 cytokine comprises the sequence of SEQ ID NO: 25.
- the signaling domain is connected to the cytokine via a cleavage sequence.
- the cleavage sequence comprises sequence elements 2A or 2S-like sequence elements.
- the anti-BCMA CAR of the present invention comprises the cleavage sequence comprising equine rhinitis virus (E2A), foot-and-mouth disease virus (F2A), Thosea asigna virus (T2A), porcine Teschovirus-1 (P2A) or a combination thereof.
- the cleavage sequence comprises T2A.
- the T2A cleavage sequence comprises the sequence of SEQ ID NO: 27.
- the cleavage sequence comprises P2A.
- the P2A cleavage sequence comprises the sequence of SEQ ID NO: 29.
- the cleavage sequence comprises E2A.
- the E2A cleavage sequence comprises the sequence of SEQ ID NO: 31.
- the anti-BCMA CAR of the present invention comprises a CD8 signal peptide, a linker, a CD8 hinge region, a CD8 transmembrane domain, a 4-1 BB costimulatory domain, and a CD3 zeta signaling domain.
- the anti-BCMA CAR of the present invention comprises a CD8 signal peptide, a linker, a CD8 hinge region, a CD8 transmembrane domain, a 4-1 BB costimulatory domain, a CD3 zeta signaling domain, a T2A cleavage sequence, and IL-15.
- the anti-BCMA CAR of the present invention comprises a CD8 signal peptide, a linker, a CD8 hinge region, a CD8 transmembrane domain, a 2B4 costimulatory domain, a CD3 zeta signaling domain, a T2A cleavage sequence, and IL-15.
- the anti-BCMA CAR of the present invention comprises a combination of the sequences selected from the odd sequences of SEQ ID NOs: 1 to 31.
- the anti-BCMA CAR of the present invention comprises one of SEQ ID NOs: 33, 35 or 37. In a more preferred embodiment, the anti-BCMA CAR of the present invention comprises SEQ ID NO: 33. In a more preferred embodiment, the anti-BCMA CAR of the present invention comprises SEQ ID NO: 35. In a more preferred embodiment, the anti-BCMA CAR of the present invention comprises SEQ ID NO: 37.
- the present invention also relates to a vector comprising the polynucleotide encoding an anti-BCMA chimeric antigen receptor (CAR), wherein the CAR comprises an anti-BCMA binding domain, a transmembrane domain, and an intracellular domain comprising one or more costimulatory domains and a signaling domain, wherein the anti-BCMA binding domain comprises the light chain variable domain of SEQ ID NO: 1 and the heavy chain variable domain of SEQ ID NO: 3.
- CAR anti-BCMA chimeric antigen receptor
- the vector comprises the polynucleotide encoding the anti-BCMA CAR of the present invention, as defined herein.
- the vector comprises the polynucleotide encoding the anti-BCMA CAR comprising one of SEQ ID NOs: 34, 36 or 38.
- the polynucleotide encoding the anti-BCMA CAR of the present invention comprises SEQ ID NO: 34.
- the polynucleotide encoding the anti-BCMA CAR of the present invention comprises SEQ ID NO: 36.
- the polynucleotide encoding the anti-BCMA CAR of the present invention comprises SEQ ID NO: 38.
- the vector of the present invention is a plasmid or an expression or transfer vector.
- the vector of the present invention comprises one or more promoters.
- the one or more promoters are selected from a cytomegalovirus (CMV) promoter, a Rous sarcoma virus (RSV) promoter or a simian virus promoter. (SV40).
- the vector of the present invention comprises one or more terminators.
- the vector of the present invention comprises one or more restriction enzyme recognition sites.
- the vector of the present invention comprises one or more regulatory elements.
- the regulatory elements may comprise, but are not limited to, origins of replication, antibiotic resistance markers, polyadenylation signals, post-transcriptional regulatory elements such as those of the hepatitis B virus (HPRE) or the woodchuck hepatitis virus (WPRE), among others.
- the vector of the present invention is a viral vector.
- the vector of the present invention is a viral vector selected from, but not limited to, retroviruses, adenoviruses, and adeno-associated viruses.
- the vector of the present invention is an adenovirus.
- the vector of the present invention is an adeno-associated virus.
- the vector of the present invention is a retrovirus.
- the vector of the present invention is a retrovirus selected from, but not limited to, a lentiviral vector and a gamma-retroviral vector.
- the vector of the present invention is a gamma-retroviral vector.
- the vector of the present invention is a lentiviral vector.
- the lentiviral vector of the present invention is selected from, but not limited to, human immunodeficiency virus (HIV) 1, human immunodeficiency virus (HIV) 2, simian immunodeficiency virus (SIV), bovine immunodeficiency virus (BIV), feline immunodeficiency virus (FIV), etc.
- the vector of the present invention further comprises virus organization genes.
- the vector comprises one or more genes encoding structural proteins, enzymes required for replication, viral envelope proteins, accessory proteins, regulatory proteins, and combinations thereof.
- the vector of the present invention also comprises the Kozak sequence (gccacc).
- the vector of the present invention corresponds to a first generation lentiviral vector in which the HIV components are separated into three plasmids.
- the vector of the present invention corresponds to a second generation lentiviral vector in which the HIV components are separated into three plasmids and the accessory genes vif, vpr, vpu, and nef have been removed.
- the vector of the present invention corresponds to a third generation lentiviral vector in which the HIV components are separated into four plasmids, the vif, vpr, vpu, nef, and tat genes have been removed, and the 5' LTR portion of the transfer plasmid has been replaced by a strong promoter.
- the vector of the present invention is a second generation lentiviral vector comprising:
- the anti-BCMA CAR expression/transfer vector of the present invention comprises a first, second, third or fourth generation vector.
- the anti-BCMA CAR expression vector of the present invention comprises a second or fourth generation vector.
- the anti-BCMA CAR expression vector of the present invention is selected from, but not limited to, plasmid p4BC, plasmid pB4CI, among others known in the art, which can be designed and synthesized according to practices common to the person skilled in the art.
- the anti-BCMA CAR expression vector of the present invention comprises the polynucleotide encoding the light and heavy chain variable domains of clone 11 D5.3.
- the anti-BCMA CAR expression vector of the present invention comprises the polynucleotide as described herein.
- the anti-BCMA CAR expression vector of the present invention comprises the nucleotide sequences of SEQ ID NOs: 2 and 4.
- the anti-BCMA CAR expression vector of the present invention comprises a combination of two or more nucleotide sequences selected from the even-numbered sequences of SEQ ID NOs: 2 to 32.
- the anti-BCMA CAR expression vector of the present invention comprises any one of SEQ ID NOs: 34, 36 or 38.
- the anti-BCMA CAR expression vector of the present invention further comprises one or more elements common to expression vectors such as one or more promoters, one or more origins of replication, one or more terminators, one or more antibiotic markers, one or more restriction enzyme recognition sites, one or more polyadenylation signals, among others.
- the anti-BCMA CAR expression vector of the present invention further comprises one or more elements common to expression vectors of SEQ ID Nos: 39 to 53.
- the anti-BCMA CAR expression vector of the present invention comprises any of the vectors as illustrated in Figures 1 a, 1 b and 1 c.
- the packaging cassette plasmid of the present invention is, but is not limited to, the psPAX2 plasmid (Addgene, #12260), which contains the lentiviral gag, pol and RRE genes.
- the psPAX2 vector is depicted in Figure 2.
- the envelope cassette plasmid is, but is not limited to, plasmid pMD2.G (Addgene, #12259), which contains the envelope coding sequences pseudotyped to the VSV-G virus.
- pMD2.G vector is depicted in Figure 3.
- the present invention also relates to a composition comprising an effector immune cell comprising the polynucleotide or vector of the present invention as described herein.
- the composition of the present invention is a pharmaceutical composition.
- pharmaceutical composition designates a preparation that is in a form that allows the biological activity of an active ingredient contained therein to be effective.
- the composition of the present invention comprises an effector immune cell comprising the polynucleotide encoding an anti-BCMA chimeric antigen receptor (CAR), wherein the CAR comprises an anti-BCMA binding domain, a transmembrane domain and an intracellular domain comprising one or more costimulatory domains and a signaling domain, wherein the anti-BCMA binding domain comprises the light chain variable domain of SEQ ID NO: 1 and the heavy chain variable domain of SEQ ID NO: 3, encoded by the nucleotide sequences of SEQ ID Nos: 2 and 4, respectively, and degenerate sequences thereof.
- CAR anti-BCMA chimeric antigen receptor
- the composition of the present invention comprises an effector immune cell comprising the vector.
- the vector comprises an expression vector comprising the polynucleotide encoding the anti-BCMA CAR comprising one of SEQ ID NOs: 34, 36 or 38.
- the polynucleotide encoding the anti-BCMA CAR of the present invention comprises SEQ ID NO: 34.
- the polynucleotide encoding the anti-BCMA CAR of the present invention comprises SEQ ID NO: 36.
- the polynucleotide encoding the anti-BCMA CAR of the present invention comprises SEQ ID NO: 38.
- the effector immune cell is transduced with the vector of the present invention.
- the effector immune cell comprises, but is not limited to, T lymphocytes, NK cells, or a combination thereof.
- the effector immune cell comprises T lymphocytes.
- the effector immune cell comprises NK cells.
- the effector immune cell comprises both T lymphocytes and NK cells.
- the effector immune cell is produced by the method of the present invention.
- the composition of the present invention further comprises one or more excipients, carriers or diluents.
- the excipients, carriers or diluents are pharmaceutically acceptable excipients known in the art, including, but not limited to, adjuvants, carriers, excipients, glidants, sweetening agents, diluents, preservatives, colorants, flavor enhancers, surfactants, wetting agents, dispersing agents, suspending agents, stabilizers, solvents, emulsifiers acceptable for use in humans or animals.
- the one or more excipients, carriers or diluents comprise sugars, starches, cellulose and its derivatives, gelatin, talc, cocoa butter, waxes, animal and vegetable fats, paraffins, silicones, bentonites, silicic acid, zinc oxide, oils, glycols, polyols, esters, agar; buffering agents, alginic acid, water, saline, Ringer's solution, alcohols, phosphate buffer solutions, and any other compatible substances used in pharmaceutical compositions.
- Those skilled in the art are highly capable of determining the pharmaceutically acceptable excipients, carriers and diluents for each composition and purpose.
- the composition of the present invention comprises an amount of CAR-expressing immune effector cells contemplated herein.
- amount refers to "an effective amount” or "a therapeutically effective amount” of a genetically modified cell, e.g., T or NK cell, to achieve a prophylactic or therapeutic result.
- a genetically modified cell e.g., T or NK cell
- composition of the present invention may comprise additional agents, such as, but not limited to, cytokines, growth factors, hormones, chemotherapeutics, radiotherapeutics, prodrugs, drugs, antibodies, etc.
- the present invention relates to a method for producing a modified effector immune cell, comprising introducing into the effector immune cell the polynucleotide or vector of the present invention, as described herein.
- the method of the present invention comprises introducing into the effector immune cell the polynucleotide encoding a anti-BCMA chimeric antigen receptor (CAR), wherein the CAR comprises an anti-BCMA binding domain, a transmembrane domain, and an intracellular domain comprising one or more costimulatory domains and a signaling domain, wherein the anti-BCMA binding domain comprises the light chain variable domain of SEQ ID NO: 1 and the heavy chain variable domain of SEQ ID NO: 3, encoded by the nucleotide sequences of SEQ ID Nos: 2 and 4, respectively, and degenerate sequences thereof.
- CAR anti-BCMA chimeric antigen receptor
- the method of the present invention comprises introducing into the effector immune cell the vector.
- the vector comprises an expression vector comprising the polynucleotide encoding the anti-BCMA CAR comprising one of SEQ ID NOs: 34, 36 or 38.
- the polynucleotide encoding the anti-BCMA CAR of the present invention comprises SEQ ID NO: 34.
- the polynucleotide encoding the anti-BCMA CAR of the present invention comprises SEQ ID NO: 36.
- the polynucleotide encoding the anti-BCMA CAR of the present invention comprises SEQ ID NO: 38.
- the effector immune cell comprises, but is not limited to, T lymphocytes, NK cells, or a combination thereof.
- the effector immune cell comprises T lymphocytes.
- the effector immune cell comprises NK cells.
- the effector immune cell comprises both T lymphocytes and NK cells.
- the effector immune cell is transduced with the vector of the present invention.
- the vector of the present invention is a lentiviral vector.
- the method of the present invention comprises, prior to the introduction of the polynucleotide or vector of the present invention into the effector immune cell, the transient transfection of the expression vector and the plasmids packaging and envelope cassettes for the production of viral particles.
- transient transfection is done with HEK (Human Embryonic Kidney) cells.
- the packaging HEK cells comprise HEK293T.
- the expression vector and the packaging and envelope cassette plasmids are mixed and dripped onto HEK293T cells, which will produce viral particles comprising the polynucleotides of the present invention, wherein the viral particles produced correspond to the vector of the present invention.
- the viral particles will be used for transduction of the effector immune cell of interest.
- the effector immune cell used in the method of the present invention comprises, but is not limited to, T lymphocytes, NK cells, or a combination thereof.
- the effector immune cell comprises T lymphocytes.
- the effector immune cell comprises NK cells.
- the effector immune cell comprises both T lymphocytes and NK cells.
- the source of the immune effector cells used in the method of the present invention is selected from, but not limited to, peripheral blood or umbilical cord blood.
- the method of the present invention comprises, prior to the introduction of the polynucleotide or vector of the present invention into the effector immune cell, the steps of collecting, isolating, selecting and activating the effector immune cells.
- the isolation of the immune effector cells is performed by concentration gradient.
- Other techniques for isolating the immune effector cells of interest are known in the art and can be used in an alternative way to those described in the present invention.
- selection of immune effector cells is performed through magnetic selection using commercially available kits or beads.
- activation of immune effector cells is performed by means of Dynabeads CD3/CD28 or interleukins appropriate for each cell type, for example, such as interleukins 12, 15 and 18.
- the method of the present invention further comprises a step of identifying the immune effector cells, which can be performed before and/or after the introduction of the polynucleotide or the vector.
- the identification of the immune effector cells is performed by immunophenotyping. Other techniques for identifying the immune effector cells are known and can be used in an alternative manner to that described in the present invention.
- the assessment of transduction efficiency is performed using commercial kits that detect CARs directed to the BCMA antigen, followed by flow cytometry. Other techniques are widely known and can be used as alternatives.
- NK cells can be obtained from autologous or allogeneic sources that include peripheral blood, bone marrow, human embryonic stem cells, induced pluripotent stem cells, umbilical cord blood or readily available NK cell lines such as NK92.
- the NK cells used in the present invention are obtained from peripheral blood.
- the NK cells used in the present invention are derived from umbilical cord (CB).
- umbilical cord blood is a readily available allogeneic source that is easily obtained and does not require healthy donors to undergo the risks of collection.
- the frequencies of NK cells in CB are ⁇ 15-20%.
- cell viability refers to the analysis of metabolically active cells in a cell culture and/or sample in order to assess their activity qualitatively and/or quantitatively. Several techniques for determining cell viability are known in the art and may be used alternatively or concomitantly.
- lymphocyte expansion corresponds to the clonal expansion of lymphocytes. Clonal expansion begins with the activation of lymphocytes through the presentation of the antigen of interest by means of the major histocompatibility complex (MHC II) molecules of APCs to T lymphocyte receptors (TCRs). This presentation promoted by APCs is called “stimulation”.
- MHC II major histocompatibility complex
- TCRs T lymphocyte receptors
- Antigen recognition allows the interaction of costimulatory molecule complexes and the secretion of cytokines, which in turn ensure the survival of antigen-specific T cells and activate factors that promote the exposure of specific gene segments, leading to the cellular differentiation of lymphocytes subdivided into subtypes (Th1, Th2 and Th17), which can be identified according to the cytokine they produce the most (INF- ⁇ , IL-4 and IL-21, respectively).
- effector immune cell corresponds to cells of the immune system that respond to their target cells without the need for co-stimulation, being normally recruited from the circulation, activated by antigens presented by macrophages and secreting cytokines.
- the medicament further comprises one or more excipients, carriers or diluents.
- the excipients, carriers or diluents are pharmaceutically acceptable ones known in the art, including, but not limited to, adjuvants, carriers, excipients, glidants, sweetening agents, diluents, preservatives, colorants, flavor enhancers, surfactants, wetting agents, dispersing agents, suspending agents, stabilizers, solvents, emulsifiers acceptable for use in humans or animals.
- the one or more excipients, carriers or diluents comprise sugars, starches, cellulose and its derivatives, gelatin, talc, cocoa butter, waxes, animal and vegetable fats, paraffins, silicones, bentonites, silicic acid, zinc oxide, oils, glycols, polyols, esters, agar; buffering agents, alginic acid, water, saline, Ringer's solution, alcohols, phosphate buffer solutions, and any other compatible substances used in pharmaceutical compositions. Professionals in the field are highly capable of determining the pharmaceutically acceptable excipients, carriers, and diluents for each drug and purpose.
- the polynucleotide, the chimeric antigen receptor, the vector, the effector immune cell produced by the method or composition, and the one or more additional agents can be administered together or sequentially.
- the present invention relates to a method for producing a modified effector immune cell, comprising introducing into the effector immune cell the polynucleotide or vector of the present invention, as described herein.
- the medicament may comprise one or more additional therapeutic agents.
- the viral vector suitable for transducing the CAR transgene of the present invention into immune effector cells is selected from, but not limited to, retroviruses, adenoviruses, and adeno-associated viruses.
- the vector of the present invention is an adenovirus.
- the vector of the present invention is an adeno-associated virus.
- the vector of the present invention is a retrovirus.
- the vector of the present invention is a retrovirus selected from, but not limited to, a lentiviral vector and a gamma-retroviral vector.
- the vector of the present invention is a gamma-retroviral vector.
- the vector of the present invention is a lentiviral vector.
- the lentiviral vector of the present invention is selected from, but not limited to, the human immunodeficiency virus (HIV).
- HAV human immunodeficiency virus
- Other viral vectors are known in the art and can be used as alternatives to the technique described herein.
- transfection refers to the process of introducing nucleic acids into eukaryotic cells.
- Cells can be transfected in a stable manner for the integration of DNA into their genome or in a transient manner for protein expression of temporary duration.
- maximum confluence corresponds to the maximum percentage of the area of the growth medium covered by adherent cells.
- the maximum confluence is about 80% to about 100%, preferably about 85%.
- immunophenotyping refers to the technique laser that evaluates cellular characteristics.
- immunophenotyping is performed by flow cytometry.
- Other cellular immunophenotyping techniques are known in the art and can be used as an alternative to the technique described here.
- Flow cytometry is a technology used to detect and measure physical and chemical aspects of particles.
- the “gating” strategy corresponds to the basic principle for analyzing the diversity of information provided by the hypotheses tested in flow cytometry, through the refinement and sequential identification of the cell populations under investigation.
- HEK-293T cells (ATCC, CRL-3216) were grown in T75 bottles (Corning, #43064111) with DMEM/F12 medium (Gibco, #11320-033) supplemented with 10% Heat-inactivated Fetal Bovine Serum (HFBS) (Gibco, #16140-071), 1X L-glutamine (Gibco, #25030-081), and 1% penicillin/streptomycin (Gibco, 15070-063) until they reached a maximum confluence of 85% in less than ten passages.
- HFBS Heat-inactivated Fetal Bovine Serum
- the plasmid DNA cocktail was prepared in 15 mL conical tubes by adding the following components: in a first tube, 6.92 ug of the plasmid psPAX2 (Addgene, #12260), 3.46 ug of the pMD2.G plasmid (Addgene, #12259), 9.62 ⁇ g of the transfer plasmid comprising one of the polynucleotides of the present invention comprising SEQ ID Nos: 34, 36 or 38, and 35 ⁇ L of P3000 to a final volume of 1.5 mL of Opti-MEM (Gibco, 31985070) (second generation vectors were used).
- the medium was discarded and 10 mL of Opti-MEM medium supplemented with 5% SFBi and 1% penicillin/streptomycin were added, and the plate was again incubated in an oven for 48 hours. After this period, the supernatant containing the viral particles was collected, centrifuged at 300 x g for 5 minutes at 4 °C and filtered through a PES filter (Microlab Scientific, #S33PES045S) with 0.45 ⁇ m pores to remove particles and cellular debris from the supernatant containing the lentiviruses. The viral supernatant was aliquoted and stored at -80 °C until transduction of T lymphocytes or NK cells.
- PES filter Microlab Scientific, #S33PES045S
- Peripheral blood collection was performed by venipuncture by the specialized team at Hospital Israelita Albert Einstein. Three tubes of blood (8.8 mL/tube) were collected in a tube with S-Monovette ACD-A (Sarstedt®, NC0504898), to obtain peripheral blood mononuclear cells (PBMC). Peripheral Blood Mononuclear Cell). All samples were alphanumerically identified prior to processing at IIEP to preserve volunteer identification.
- Peripheral blood mononuclear cells were obtained using a Ficoll-Paque concentration gradient (Cytiva, #17144003), which separates mononuclear cells from red blood cells and plasma.
- Blood diluted 1:1 with 1X PBS was added to a conical tube containing Ficoll-Paque, the tubes were centrifuged at 400 x g for 40 min with acceleration set to 4 and deceleration set to 0, and the interphase containing PBMCs was collected and washed with phosphate-buffered saline (PBS) (Gibco, #10010-031).
- PBS phosphate-buffered saline
- T cell selection from PBMC was performed by magnetic selection using the Pan T Cell Isolation Kit (Miltenyi, 130-096-535) and the LD column (Miltenyi, 130-042-901), following the manufacturer's instructions.
- Activation of the selected T cells was performed with anti-CD3/CD28 magnetic beads (Dynabeads, Gibco, 11161 D) at a ratio of 1 bead : 1 cell. The selection and activation steps are also discussed below.
- the selected cells were activated with CD3/CD28 Dynabeads for in vitro maintenance and expansion of the T lymphocyte population. Twenty-four hours after addition of the activation Dynabeads, the activation markers CD25 (BD Pharmigen, 555432) and CD69 (BD Biosciences, #340560) were identified by cytometry ( Figure 6).
- FIG. 6 shows that, before activation with Dynabeads, 94.2% of the cells did not express any of the activation markers. Already 24 hours after activation, approximately 90% of the cells showed expression of at least one activation marker, and almost 85% showed expression of both markers. Thus, it can be concluded that the lymphocytes were properly activated.
- CAR-T cells For the production of CAR-T cells, cells were transduced with previously produced lentiviral vectors (e.g., see Example 1). For this, 2.5 x 10 5 activated T cells were seeded per well in 24-well plates in supplemented RPMI culture medium. Then, 1 ml of the lentivirus suspension (non-concentrated) was added. The addition of polybrene at a concentration of 8 pg/mL is considered optional. The plates were homogenized and incubated in a cell culture incubator at 37 °C, with 5% CO 2 and controlled humidity for 16 hours. The following day, the cells were washed and seeded again on the plates in supplemented RPMI. The cells were expanded, maintaining the concentration of 2 x 10 5 cells/mL up to 1 x 106 cells/mL of culture medium, with medium change every 2 days (more frequent when necessary) and transfer to larger bottles as needed.
- the cells were washed again and labeled with anti-CD3 and anti-CD56 antibodies for 15 minutes at room temperature in the dark.
- an anti-biotin secondary antibody was also added. Labeled non-transduced T cells were used as a negative control.
- Acetoxymethyl calcein is a dye used to determine the viability of eukaryotic cells. Because it is permeant, calcein passes into living cells, where it fluoresces after hydrolysis of the acetoxymethyl moiety by active intracellular esterases. Dead cells do not have active esterases and therefore do not fluoresce.
- the calcein AM cytotoxicity assay was performed using 4 ⁇ 10 5 target cells, MM1.S (ATCC, CRL-2974) or Nalm6 (ATCC, CRL-3273) lines, which were labeled with 1 ml of calcein AM (ThermoFisher, C1430) at 25 nM for 30 min in the dark. Then, the cells were washed twice with 10 mL of DPBS + 5% SFBi, resuspended in RPMI medium and 1 ⁇ 10 4 cells were plated per well in a 96-well plate (Corning, #3799).
- Figure 10 corresponds to the experimental design of the calcein assay.
- CAR-T or non-transduced lymphocytes were counted and added to the wells containing the target cells in the following ratios: 1:1, 1:2, 2:1, 5:1 and 10:1 (effector cells: target cells).
- Target cells without calcein labeling, calcein-labeled target cells, and target cells killed with 0.1% triton solution were used as positive and negative controls of the assay. All conditions were acquired by flow cytometry in triplicate.
- Effector cells and target cells were incubated for 2 to 4 h in an incubator and, after incubation, readings were taken on the Attune flow cytometer (ThermoFisher), in the channel corresponding to the FITC fluorochrome to identify live target cells (labeled with calcein-AM).
- Anti-BCMA CAR-T cells (with and without IL-15) comprising the anti-BCMA CARs of SEQ ID NO: 33 and 35 were co-cultured with MM1 S target cells for two hours and the rate of target cell killing was analyzed. CAR-T cells were also co-cultured with the Nalm6 cell line to verify the specificity of the lymphocyte activity. In addition, co-cultures were performed between non-transduced lymphocytes to verify the basal cytotoxicity of T lymphocytes against the tested cell lines, and between anti-CD19 CAR-T cells to verify the cytotoxicity of genetically modified T lymphocytes in a non-BCMA-specific and CD19-specific manner (Figure 11).
- the anti-BCMA CAR-T lymphocytes of the present invention were more capable of killing MM1.S lineage cells than Nalm6 cells ( Figure 11 B), showing the specificity of the cytotoxic action to the BCMA antigen.
- Figure 11 B The analysis of the different proportions of effector cells to target cells allows us to observe that there was a significant increase in the death rates obtained between the smaller proportions (0.5:1, 1:1 and 2:1), however this increase was milder when analyzing the larger proportions (5:1 and 10:1), evidencing a potential plateau in the death potential of the cells.
- the cytokine production assay is performed with the aim of identifying the production of the cytokines IFN-y and TNF-a in the anti-BCMA CAR-T lymphocytes comprising the anti-BCMA CAR of SEQ ID NO: 33 and 35, when they were stimulated. For this, four experimental conditions were tested:
- lymphocytes were stimulated with 1X Stimulation Cocktail containing PMA and lonomycin (Invitrogen, #00-4970-93);
- the cells were passed into cytometry tubes, the wells were washed with 300 pL of FACS Buffer and the contents were added to the respective tubes. The cells were then labeled for CAR receptor identification with the CAR BCMA Detection reagent (Creative BioLabs), washed and then labeled with 1 pL of LIVE/DEAD (InvitrogenTM) for 20 minutes. The cells were washed again and incubated with 10 pL and 1.25 pL of the anti-CD3 antibodies. and anti-CD56, respectively, for 15 minutes.
- CAR BCMA Detection reagent Creative BioLabs
- LIVE/DEAD InvitrogenTM
- ML cells For activation of ML cells, they were seeded following the proportion of 1.0 X 106 cells/mL in a 24-well plate (Corning, #3473), adding the interleukins IL-12 (10 ng/mL), IL-15 (50 ng/mL) and IL-18 (50 ng/mL) and incubating for 16 hours.
- Immunophenotyping was performed using 5.0 x 105 cells per tube using the flow cytometry technique, using a panel with 3 markers (Table 3).
- the transduction process consisted of adding 1 mL of the unconcentrated virus suspension to 24-well plates previously treated with 1 pg/mL of RetroNectin (Takara, T100B) and incubating for 4 hours in an oven at 37 °C, 5% CO2 atmosphere. After the incubation time, 2.5 x 10 5 of NK or ML cells were seeded per well using supplemented RPMI 1640+Click's medium, centrifugation was performed at 1,000 xg for 1 hour at 32 °C and 10% SFBi was added. The following day (D6), the culture medium was changed and feeder cells were added in a 1:1 ratio (feeder: NK).
- Figure 15 exemplifies the gating strategy used for analysis of NK cells expressing the anti-BCMA-IL-15 CAR comprising SEQ ID NO: 37.
- Transduced and non-transduced cells were maintained in culture for 21 days and monitored for expansion rate and viability.
- acetoxymethyl calcein passes into living cells, where it fluoresces following hydrolysis of the acetoxymethyl moiety mediated by active intracellular esterases. Dead cells do not have active esterases and therefore do not fluoresce.
- CAR-NK, CAR-NKML or the respective non-transduced groups were counted and added to the wells containing the target cells at the ratios of 0.5:1, 1:1, 2:1, 5:1 and 10:1 (effector cells:target cells).
- Target cells without calcein labeling, calcein-labeled target cells and target cells killed with 0.1% triton solution were used as positive and negative controls of the assay. All conditions were acquired by flow cytometry in triplicate.
- Effector cells and target cells were incubated for 4 h in an incubator at 37 °C in a CO2 atmosphere and, after the incubation time, acquisition was performed on the Attune flow cytometer, in the channel corresponding to the FITC fluorochrome to identify live target cells (labeled with calcein-AM).
- the cell death rate graphs for NK and ML cells transduced with the anti-BCMA CAR of the present invention comprising SEQ ID NO: 37, encoded by SEQ ID NO: 38, and non-transduced can be observed.
- the transduced NK and ML cells promoted a higher death rate than the respective non-transduced cells (described in the graphs as NT), with P ⁇ 0.0001 for the comparison in both groups (comparison between transduced NK and non-transduced NK and transduced ML and non-transduced ML).
- the difference in the death rate between the transduced and non-transduced groups was 30%.
- the cytokine production assay was performed with the aim of identifying the production of the cytokines IFN-y and TNF-a and the expression of CD107a in anti-BCMA CAR-NK and CAR-NKML cells, when these were stimulated in the presence of the target cell MM1 S.
- the experiment consisted of three experimental conditions:
- a 96-well plate 1.5 x10 5 of anti-BCMA CAR-NK or CAR-NKML cells of the present invention, in which the CAR comprises SEQ ID NO: 37, or their respective non-transduced groups, 0.2 pL of Brefeldin A (Thermo Fisher) and 0.75 pL of the anti-CD107a antibody (BV785, BD Biosciences) were added.
- 1X of the stimulation cocktail Invitrogen was added and in the stimulation wells with target cells, 1.5 x 10 5 MM1 S cells were added per well. The conditions were performed in triplicate, with a final volume of 200 pL per well and the plate was incubated in an oven at 37°C, with a CO 2 atmosphere for 6 hours.
- the cells were transferred from the 96-well plate to cytometry tubes, and each experimental well was washed with 300 pL of FACS Buffer. After being transferred to the cytometry tube, the cells were labeled for CAR receptor identification with the CAR BCMA Detection reagent (Creative BioLabs) for 45 minutes, washed and labeled with 1 pL of LIVE/DEAD (InvitrogenTM) for 20 minutes.
- the cells were then fixed and permeabilized using the Fixation/Permeabilization Kit (BD Biosciences, 554714) and intracellular labeling of anti-IFN-y (PE, BD Biosciences) and anti-TNF-a (PE-Cy7, Biolegend) cytokines was performed using 2.5 pL of antibody per sample. Data acquisition was performed on the Attune Nxt flow cytometer (Thermo Fisher Scientific) and analysis was performed using FlowJo Software v10.6.0. The antibody panel is indicated below. used (Table 4) and the gating strategy used for the analysis ( Figure 19).
- T-cell receptor (TCR) activation An enduring enigma. J Biol Chem. 2020; 295(4):914-25.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Genetics & Genomics (AREA)
- Organic Chemistry (AREA)
- Engineering & Computer Science (AREA)
- Biomedical Technology (AREA)
- Zoology (AREA)
- Wood Science & Technology (AREA)
- General Health & Medical Sciences (AREA)
- Biotechnology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Biochemistry (AREA)
- General Engineering & Computer Science (AREA)
- Molecular Biology (AREA)
- Biophysics (AREA)
- Microbiology (AREA)
- Immunology (AREA)
- Medicinal Chemistry (AREA)
- Plant Pathology (AREA)
- Physics & Mathematics (AREA)
- Cell Biology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Toxicology (AREA)
- Gastroenterology & Hepatology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Peptides Or Proteins (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Abstract
Description
POLINUCLEOTÍDEO, RECEPTOR DE ANTÍGENO QUIMÉRICO, VETOR, COMPOSIÇÃO, MÉTODO PARA PRODUÇÃO DE CÉLULA IMUNE EFETORA MODIFICADA E USO DO POLINUCLEOTÍDEOPOLYNUCLEOTIDE, CHIMERIC ANTIGEN RECEPTOR, VECTOR, COMPOSITION, METHOD FOR PRODUCTION OF MODIFIED EFFECTOR IMMUNE CELL AND USE OF POLYNUCLEOTIDE
CAMPO DA INVENÇÃO FIELD OF INVENTION
A presente invenção está relacionada a um polinucleotídeo que codifica um receptor de antígeno quimérico (CAR) anti-BCMA e ao polipeptídeo que corresponde ao receptor de antígeno quimérico (CAR) anti-BCMA por si. A presente invenção também está relacionada a um vetor e uma composição, compreendendo uma célula imune efetora, que compreendem o polinucleotídeo, além de um método para a produção de célula imune efetora modificada e do uso do polinucleotídeo, do vetor, da composição ou da célula imune efetora produzida pelo método para a fabricação de um medicamento para o tratamento de mieloma múltiplo. The present invention relates to a polynucleotide encoding an anti-BCMA chimeric antigen receptor (CAR) and the polypeptide corresponding to the anti-BCMA chimeric antigen receptor (CAR) itself. The present invention also relates to a vector and a composition comprising an effector immune cell comprising the polynucleotide, in addition to a method for producing a modified effector immune cell and the use of the polynucleotide, the vector, the composition or the effector immune cell produced by the method for the manufacture of a medicament for the treatment of multiple myeloma.
ANTECEDENTES DA INVENÇÃO BACKGROUND OF THE INVENTION
O mieloma múltiplo (MM) é um tipo de câncer hematológico que compromete as células plasmáticas na medula óssea (MO) e a doença é caracterizada pela proliferação de células plasmáticas malignas, que produzem imunoglobulinas monoclonais defeituosas e/ou em excesso, mesmo na ausência de antígenos estimulantes, resultando em disfunções em múltiplos órgãos (Palumbo A., 2011 ; Feng, D., 2020). Multiple myeloma (MM) is a type of hematologic cancer that affects plasma cells in the bone marrow (BM) and the disease is characterized by the proliferation of malignant plasma cells, which produce defective and/or excess monoclonal immunoglobulins, even in the absence of stimulating antigens, resulting in dysfunctions in multiple organs (Palumbo A., 2011; Feng, D., 2020).
A maior parte dos pacientes com MM inicialmente apresentam a gamopatia monoclonal de significado indeterminado (MGUS), caracterizada pela presença da proteína monoclonal (proteína M) no sangue. Um a dois porcento dos casos de MGUS progridem para casos de MM por ano (Atkin et al., 2018; Kyle et al., 2018). O estágio intermediário entre MGUS e MM é caracterizado como mieloma múltiplo latente (SMM). Neste estágio, os pacientes apresentam maiores alterações da proteína M sanguínea e contagem de células plasmáticas clonais, mas ainda não apresentam sintomas. Cerca de 10% dos pacientes progridem para MM por ano (Ravindran et al., 2016). O diagnóstico do mieloma múltiplo é definido quando é observado um ou mais sintomas que incluem hipercalcemia, falência renal, anemia, elevação da proteína M no soro e lesões ósseas líticas, e 10% ou mais de células plasmáticas na medula óssea ou plasmocitoma identificadas por biópsia (Rajkumar et al., 2014; Rajkumar, S.V., 2022). Most patients with MM initially present with monoclonal gammopathy of undetermined significance (MGUS), characterized by the presence of monoclonal protein (M protein) in the blood. One to two percent of MGUS cases progress to MM per year (Atkin et al., 2018; Kyle et al., 2018). The intermediate stage between MGUS and MM is characterized as smoldering multiple myeloma (SMM). At this stage, patients have greater changes in blood M protein and clonal plasma cell counts but do not yet have symptoms. Approximately 10% of patients progress to MM per year (Ravindran et al., 2016). The diagnosis of multiple myeloma is defined when one or more symptoms are observed, including hypercalcemia, renal failure, anemia, elevated serum M protein and lytic bone lesions, and 10% or more plasma cells in the bone marrow or plasmacytoma identified by biopsy (Rajkumar et al., 2014; Rajkumar, SV, 2022).
De acordo com as estatísticas da Global Cancer Observatory (GCO), houve, em 2018, cerca de 160.000 casos de mieloma múltiplo mundialmente, o que representa aproximadamente 1 % das doenças neoplásicas e 13% dos cânceres hematológicos. A população jovem é raramente acometida, sendo a média da idade de 70 anos ao diagnóstico. No Brasil, foi verificada a incidência de cerca de 3.500 casos em 2012, e é estimado que cerca de 30.000 pacientes com mieloma múltiplo estejam sob tratamento (da Silva Ferreira et al., 2017; Padala et al., 2021 ). According to statistics from the Global Cancer Observatory (GCO), in 2018 there were approximately 160,000 cases of multiple myeloma worldwide, representing approximately 1% of neoplastic diseases and 13% of hematologic cancers. The young population is rarely affected, with the average age at diagnosis being 70 years. In Brazil, the incidence of approximately 3,500 cases was recorded in 2012, and it is estimated that approximately 30,000 patients with multiple myeloma are undergoing treatment (da Silva Ferreira et al., 2017; Padala et al., 2021).
Porém, o mieloma múltiplo é atualmente considerado como uma doença incurável. Sendo assim, o objetivo primário do tratamento é o aumento da sobrevida e qualidade de vida dos pacientes. As opções de tratamento visam diminuir a abundância de células plasmáticas malignas na MO e incluem procedimentos como: transplante de células tronco hematopoiéticas (TCTH), radioterapia e quimioterapia. Na última década, o surgimento de novas opções de tratamento, como inibidores de proteassoma de 2a geração, drogas imunomoduladoras de 2a geração e anticorpos monoclonais anti-CD38 resultaram em uma significativa melhora na sobrevida dos pacientes com mieloma múltiplo (Cowan et al., 2022; Rodríguez-Otero et al., 2020). However, multiple myeloma is currently considered an incurable disease. Therefore, the primary goal of treatment is to increase survival and quality of life of patients. Treatment options aim to reduce the abundance of malignant plasma cells in BM and include procedures such as hematopoietic stem cell transplantation (HSCT), radiotherapy, and chemotherapy. In the last decade, the emergence of new treatment options, such as 2nd generation proteasome inhibitors, 2nd generation immunomodulatory drugs, and anti-CD38 monoclonal antibodies, have resulted in a significant improvement in the survival of patients with multiple myeloma (Cowan et al., 2022; Rodríguez-Otero et al., 2020).
Entretanto, a doença tem um perfil recidivante e progressivo, levando à refratariedade ou recidiva na maioria dos casos. Nestes casos, o tratamento do mieloma múltiplo tem como principal objetivo prolongar a sobrevida dos pacientes e o tempo até a progressão da doença. As opções de tratamento do MM recidivado ou refratário englobam anticorpos monoclonais anti-CD38 ou anti-CD319, esteroides, drogas imunomoduladoras, inibidores de proteassoma, agentes alquilantes, agentes antimetabólicos e transplante (Cowan et al., 2022; Shah et al., 2012). O tratamento do mieloma múltiplo gera altos gastos para os sistemas e operadoras de saúde do país e do mundo, pois o manejo da doença é de alto custo. Estima-se que o custo anual no Brasil seja de R$124.144 por paciente, valor referente aos custos com internação, medicamentos e manejo de complicações (Rodríguez- Otero et al., 2020; Pepe et al., 2018). However, the disease has a relapsing and progressive profile, leading to refractoriness or relapse in most cases. In these cases, the main objective of treating multiple myeloma is to prolong patient survival and the time until disease progression. Treatment options for relapsed or refractory MM include anti-CD38 or anti-CD319 monoclonal antibodies, steroids, immunomodulatory drugs, proteasome inhibitors, alkylating agents, antimetabolic agents, and transplantation (Cowan et al., 2022; Shah et al., 2012). The treatment of multiple myeloma generates high costs for health systems and operators in the country and worldwide, as the management of the disease is expensive. It is estimated that the annual cost in Brazil is R$124,144 per patient, a value referring to the costs of hospitalization, medications and management of complications (Rodríguez-Otero et al., 2020; Pepe et al., 2018).
Diante do cenário atual, observa-se uma clara necessidade de novas opções de tratamento para os pacientes com mieloma múltiplo, principalmente em casos de refratariedade e recidivas, em que, muitas vezes, a maior parte dos tratamentos disponíveis já foram utilizados. Neste contexto, a imunoterapia, como a utilização de células CAR-T e/ou CAR-NK pode fornecer grande ajuda em diversos aspectos (Kõhler et al., 2018; Perez-Amill et al., 2021 ). Given the current scenario, there is a clear need for new treatment options for patients with multiple myeloma, especially in cases of refractoriness and relapses, in which, often, most of the available treatments have already been used. In this context, immunotherapy, such as the use of CAR-T and/or CAR-NK cells, can provide great help in several aspects (Kõhler et al., 2018; Perez-Amill et al., 2021).
De maneira geral, um receptor CAR (do inglês - chimeric antigen receptor) é constituído de três domínios ligados entre si, sendo eles: um domínio de reconhecimento extracelular, um domínio transmembrana e domínios de sinalização intracelulares (também conhecido como domínio citoplasmático). O domínio extracelular é formado por porções variáveis das cadeias leves e pesadas de uma imunoglobulina específicas para o reconhecimento de algum antígeno de escolha, opcionalmente ligadas por um ligante flexível (Ramos, C.A., Dotti, G., 2011 ). In general, a CAR receptor (chimeric antigen receptor) consists of three linked domains: an extracellular recognition domain, a transmembrane domain, and intracellular signaling domains (also known as the cytoplasmic domain). The extracellular domain is formed by variable portions of the light and heavy chains of an immunoglobulin specific for the recognition of an antigen of choice, optionally linked by a flexible linker (Ramos, C.A., Dotti, G., 2011).
Esta porção extracelular é ligada a um domínio transmembrana que, por sua vez, liga-se ao domínio de sinalização intracelular. A ligação entre o domínio extracelular e o domínio transmembrana é denominada hinge (dobradiça) (Miliotou, A.N., Papadopoulou, L.C., 2018). O domínio intracelular pode combinar domínios sinalizadores do complexo de receptores TCR e de moléculas coestimulatórias dos linfócitos T. Estas moléculas coestimulatórias são necessárias para aumentar a proliferação, citotoxicidade e persistência destas células in vitro e in vivo. Assim, a terapia com células CAR combina a alta especificidade de anticorpos monoclonais com a citotoxicidade potente e a persistência a longo prazo de células T citotóxicas (Barrett et al., 2014; Zhang et al., 2017). A terapia com células CAR-T é um tipo de tratamento em que as células T do paciente são geneticamente modificadas para que essas tornem-se específicas e eficazes para atacar as células cancerosas. Para isso, linfócitos T são geralmente coletados do próprio paciente e, neles, é inserido um gene que codifica um CAR. Este receptor reconhece especificamente uma proteína presente nas células tumorais do paciente, aumentando a afinidade dos linfócitos T (neste caso, denominado de CAR- T) às células tumorais. Essas células são então expandidas e infundidas no paciente, onde continuam a expandir e podem reconhecer e matar as células cancerosas (Picanto-Castro et al., 2020; Miliotou et al., 2018). This extracellular portion is linked to a transmembrane domain, which in turn binds to the intracellular signaling domain. The link between the extracellular domain and the transmembrane domain is called the hinge (Miliotou, AN, Papadopoulou, LC, 2018). The intracellular domain can combine signaling domains of the TCR receptor complex and costimulatory molecules of T lymphocytes. These costimulatory molecules are necessary to increase the proliferation, cytotoxicity and persistence of these cells in vitro and in vivo. Thus, CAR cell therapy combines the high specificity of monoclonal antibodies with the potent cytotoxicity and long-term persistence of cytotoxic T cells (Barrett et al., 2014; Zhang et al., 2017). CAR-T cell therapy is a type of treatment in which the patient's T cells are genetically modified so that they become specific and effective in attacking cancer cells. To do this, T lymphocytes are usually collected from the patient and a gene encoding a CAR is inserted into them. This receptor specifically recognizes a protein present in the patient's tumor cells, increasing the affinity of the T lymphocytes (in this case, called CAR-T) for the tumor cells. These cells are then expanded and infused into the patient, where they continue to expand and can recognize and kill cancer cells (Picanto-Castro et al., 2020; Miliotou et al., 2018).
Já na terapia com células CAR-NK, as células NK do paciente ou de outro doador são geneticamente modificadas para atacar as células cancerosas. De forma semelhante às células T, é inserido um gene que codifica um CAR nas células NK que depois são então expandidas e infundidas no paciente, onde continuam a expandir e podem reconhecer e matar as células cancerosas. In CAR-NK cell therapy, NK cells from the patient or another donor are genetically modified to attack cancer cells. Similar to T cells, a gene encoding a CAR is inserted into the NK cells, which are then expanded and infused into the patient, where they continue to expand and can recognize and kill cancer cells.
A terapia com células CAR-NK surge de forma alternativa às células CAR-T visando superar algumas limitações como (i) a dificuldade de elaboração de doses clinicamente relevantes de células CAR-T autólogas em pacientes linfopênicos pré-tratados ou (ii) o risco do desenvolvimento da doença do enxerto contra hospedeiro (DECH) atrelado ao uso de células T coletadas de uma fonte alogênica, isto é, de outro doador (Mehta et al., 2018; Goulmy, E., 1997). As células NK representam os efetores mais eficazes contra tumores com um mecanismo de ação distinto ao mecanismo das células T (Davies et al., 2014; Vivier et al., 2012). Em contraste aos outros linfócitos, as células NK não expressam receptores antígeno específicos, mas sim receptores codificados pela linha germ inativa, que são ativadores ou inibidores que podem induzir sinal positivo ou negativo, e o balanço desses sinais controlam a função efetora das células NK (Lanier, L.L., 1998). As células NK maduras também têm um tempo de vida relativamente limitado, permitindo uma atividade antitumoral eficaz, mas reduzindo simultaneamente a probabilidade de acontecimentos adversos a longo prazo, tais como citopenias prolongadas, devido ao reconhecimento em alvo não tumoral (tecidos normais), como a aplasia de células B (Maude et al., 2014). Além disso, as células CAR-NK mantêm a sua capacidade intrínseca de reconhecer as células tumorais por meio de seus receptores nativos. Dessa forma, quando comparadas às células T no tratamento com CAR, teoricamente, é menos provável que as células tumorais escapem da vigilância imunológica das células NK, mesmo que as células tumorais regulem negativamente o antígeno alvo de CAR (Mehta et al., 2018; Sotillo et al., 2015). CAR-NK cell therapy emerges as an alternative to CAR-T cells to overcome some limitations such as (i) the difficulty in preparing clinically relevant doses of autologous CAR-T cells in pretreated lymphopenic patients or (ii) the risk of developing graft-versus-host disease (GVHD) associated with the use of T cells collected from an allogeneic source, that is, from another donor (Mehta et al., 2018; Goulmy, E., 1997). NK cells represent the most effective effectors against tumors with a mechanism of action distinct from that of T cells (Davies et al., 2014; Vivier et al., 2012). In contrast to other lymphocytes, NK cells do not express antigen-specific receptors, but rather inactive germline-encoded receptors that are either activators or inhibitors that can induce either a positive or negative signal, and the balance of these signals controls the effector function of NK cells (Lanier, LL, 1998). Mature NK cells also have a relatively limited life span, allowing effective antitumor activity but simultaneously reducing the likelihood of long-term adverse events, such as prolonged cytopenias, due to recognition in non-tumor targets (normal tissues), such as B-cell aplasia (Maude et al., 2014). Furthermore, CAR-NK cells maintain their intrinsic ability to recognize tumor cells through their native receptors. Thus, when compared to T cells in CAR treatment, theoretically, tumor cells are less likely to escape the immune surveillance of NK cells, even if tumor cells downregulate the CAR target antigen (Mehta et al., 2018; Sotillo et al., 2015).
A resposta clínica da alorreatividade de células NK foi evidenciada em vários estudos no cenário de transplante de células-tronco hematopoiéticas (TCTH), em que pacientes que receberam um enxerto contendo células NK alorreativas tiveram um risco significativamente menor de recaída e melhor sobrevida (Ruggeri et al., 2002; Cooley et al., 2009; Oevermann et al., 2014; Cooley et al., 2010; Cooley et al., 2014). A transferência adotiva de células NK alorreativas como terapia autônoma (independente do TCTH) também demonstrou resultados encorajadores em uma variedade de malignidades (Miller et al., 2005; lliopoulou et al., 2010; Rubnitz et al., 2010; Curti et al., 2011 ; Geller et al., 2011 ; Bachanova et al., 2014). Já o sucesso do efeito antitumoral das células CAR-NK vem sendo confirmado em diversos estudos pré-clínicos, entretanto, ainda são poucos os estudos clínicos em vigência atualmente (Liu et al., 2018; Kerbauy et al., 2017). The clinical response of NK cell alloreactivity has been demonstrated in several studies in the setting of hematopoietic stem cell transplantation (HSCT), where patients who received a graft containing alloreactive NK cells had a significantly lower risk of relapse and improved survival (Ruggeri et al., 2002; Cooley et al., 2009; Oevermann et al., 2014; Cooley et al., 2010; Cooley et al., 2014). Adoptive transfer of alloreactive NK cells as stand-alone therapy (independent of HSCT) has also shown encouraging results in a variety of malignancies (Miller et al., 2005; Liopoulou et al., 2010; Rubnitz et al., 2010; Curti et al., 2011; Geller et al., 2011; Bachanova et al., 2014). The success of the antitumor effect of CAR-NK cells has been confirmed in several preclinical studies; however, there are still few clinical studies currently in progress (Liu et al., 2018; Kerbauy et al., 2017).
Até o momento, existem cinco gerações de receptores CAR, que diferem entre si em relação a sua estrutura e complexidade. A primeira geração é constituída pelo domínio extracelular fundido a um domínio citoplasmático CD3 zeta. Apesar de apresentarem eficácia in vitro, o mesmo não foi observado em ensaios in vivo. Os CARs de segunda geração possuem um domínio de sinalização coestimulatória adicional, enquanto os CARs de terceira geração apresentam dois ou mais destes domínios coestimulatórios. A adição de domínios coestimulatórios gera maior proliferação, citotoxicidade, persistência e eficácia clínica (Ramos, C.A., Dotti, G., 2011 ; Essand, M., Loskog, A.S.I., 2013). A adição de dois domínios coestimulatórios em CARs de 3a geração visam aumentar ainda mais a potência dos linfócitos CAR-T, porém os ensaios que compararam CARs de 2a e 3a geração mostraram resultados controversos, de modo que ainda não é comprovada a superioridade dos CARs de 3a geração em relação aos de 2a geração. Os CARs de 4a geração apresentam os componentes anteriormente listados com a adição de outros genes, como os de citocinas, com o objetivo de ampliar a atividade das células no combate ao tumor. Estes CARs, uma vez ativados, além de iniciarem mecanismos de morte contra o tumor, levam à produção e secreção de citocinas, que agem de maneira autócrina, aumentando sua ativação, e também ativando outras células do sistema imune, aumentando assim a resposta antitumoral. Por fim, os CARs de 5a geração estão sendo atualmente desenvolvidos. Eles contêm o domínio da cadeia [3 da porção citoplasmática do receptor de I L-2 ( I L-2R|3) , responsável por ativar a via JAK-STAT de maneira antígeno-dependente, levando à ativação e proliferação dos linfócitos T (Wilkins et al., 2017; Till et al., 2012). To date, there are five generations of CAR receptors, which differ from each other in terms of their structure and complexity. The first generation consists of the extracellular domain fused to a CD3 zeta cytoplasmic domain. Although they are effective in vitro, this has not been observed in in vivo assays. Second-generation CARs have an additional costimulatory signaling domain, while third-generation CARs have two or more of these costimulatory domains. The addition of costimulatory domains generates greater proliferation, cytotoxicity, persistence and clinical efficacy (Ramos, CA, Dotti, G., 2011 ; Essand, M., Loskog, ASI, 2013). The addition of two costimulatory domains in 3rd generation CARs aims to further increase the potency of CAR-T lymphocytes, but trials comparing 2nd and 3rd generation CARs have shown controversial results, so that the superiority of 3rd generation CARs over 2nd generation CARs has not yet been proven. 4th generation CARs present the components previously listed with the addition of other genes, such as cytokines, with the aim of increasing the activity of the cells in fighting the tumor. These CARs, once activated, in addition to initiating death mechanisms against the tumor, lead to the production and secretion of cytokines, which act in an autocrine manner, increasing their activation, and also activating other cells of the immune system, thus increasing the antitumor response. Finally, 5th generation CARs are currently being developed. They contain the domain of the [3 chain of the cytoplasmic portion of the IL-2 receptor (IL-2R|3), responsible for activating the JAK-STAT pathway in an antigen-dependent manner, leading to the activation and proliferation of T lymphocytes (Wilkins et al., 2017; Till et al., 2012).
A escolha do antígeno alvo da terapia com células CAR é uma etapa extremamente importante para o sucesso do tratamento. O antígeno alvo ideal deve ser fortemente expresso nas células alvo do tratamento, para que as células CAR possam facilmente localizar as células tumorais a fim de eliminá-las. Além disso, o antígeno deve ser exclusivamente expresso nas células alvo, ou ter baixa expressão em outros tipos celulares. Isto é importante para que as células CAR não exerçam sua função em tecidos saudáveis ou inespecíficos, evitando efeitos adversos (Wei et al., 2019). Choosing the target antigen for CAR cell therapy is an extremely important step for successful treatment. The ideal target antigen should be strongly expressed in the target cells of the treatment, so that the CAR cells can easily locate the tumor cells in order to eliminate them. In addition, the antigen should be exclusively expressed in the target cells, or have low expression in other cell types. This is important so that the CAR cells do not exert their function in healthy or nonspecific tissues, avoiding adverse effects (Wei et al., 2019).
No contexto do mieloma múltiplo, o antígeno de maturação das células B (BCMA) é o mais estudado (Picanco-Castro et al., 2020; Barrett et al., 2014; Hay, K.A, Turtle, CJ., 2017; Sermer, D., Brentjens, R., 2019). A proteína BCMA pertence à superfamília TNFR e tem sua expressão restrita ao compartimento de células B. Sua expressão é encontrada em células plasmáticas clonais, policlonais e uma pequena porção de células B do centro germ inativo, de memória e plasmoblastos. A expressão de BCMA nas células de pacientes com mieloma múltiplo é praticamente universal e, em vários casos, essa proteína apresenta superexpressão, o que a toma um alvo ideal para a terapia com células CAR (Rodríguez-Otero et al., 2020; Dogan et al., 2020; Fridman et al., 2018; Shah et al., 2020). In the context of multiple myeloma, B cell maturation antigen (BCMA) is the most studied (Picanco-Castro et al., 2020; Barrett et al., 2014; Hay, K. A., Turtle, C. J., 2017; Sermer, D., Brentjens, R., 2019). The BCMA protein belongs to the TNFR superfamily and its expression is restricted to the B cell compartment. Its expression is found in clonal, polyclonal plasma cells and a small portion of inactive germ center B cells, memory cells, and plasmablasts. BCMA expression in cells from patients with multiple myeloma is virtually universal, and in several cases, this protein is overexpressed, making it an ideal target for CAR cell therapy (Rodríguez-Otero et al., 2020; Dogan et al., 2020; Fridman et al., 2018; Shah et al., 2020).
Atualmente, dois produtos de células CAR-T anti-BCMA são aprovados pelo U.S. Food and Drug Administration (FDA) para uso no tratamento do mieloma múltiplo, sendo eles Idecabtagene-vicleucel e Citacabtagene-autoleucel. Este último foi também aprovado pela Agência Nacional de Vigilância Sanitária (ANVISA) em 2022 para o tratamento de pacientes com mieloma múltiplo recidivado ou refratário, que receberam anteriormente um inibidor do proteassoma, um agente imunomodulador e um anticorpo anti-CD38 (Munshi et al., 2023; Martin et al., 2023). Currently, two anti-BCMA CAR-T cell products are approved by the U.S. Food and Drug Administration (FDA) for use in the treatment of multiple myeloma, namely Idecabtagene-vicleucel and Citacabtagene-autoleucel. The latter was also approved by the Brazilian Health Regulatory Agency (ANVISA) in 2022 for the treatment of patients with relapsed or refractory multiple myeloma who have previously received a proteasome inhibitor, an immunomodulatory agent, and an anti-CD38 antibody (Munshi et al., 2023; Martin et al., 2023).
Os produtos CAR-T anti-BCMA aprovados para o tratamento do mieloma múltiplo até o momento ainda são utilizados como 5a linha de tratamento, mas apresentam potencial de utilização em linhas anteriores, estando dependente do aprimoramento das técnicas para alcançar condições ideais para o tratamento dos pacientes (Cappell, K.M., Kochenderfer, J.N., 2023). The anti-BCMA CAR-T products approved for the treatment of multiple myeloma to date are still used as the 5th line of treatment, but have potential for use in earlier lines, depending on the improvement of techniques to achieve ideal conditions for treating patients (Cappell, KM, Kochenderfer, JN, 2023).
Diversos estudos e ensaios clínicos foram e continuam sendo feitos para o desenvolvimento e aprimoramento das terapias com células CAR-T e CAR-NK. Estudos anteriores demonstraram ótima eficácia terapêutica, incentivando a continuidade do desenvolvimento dessas terapias e os estudos atuais investigam técnicas para melhorar a eficácia, diminuir a toxicidade e amenizar os efeitos adversos das mesmas (Littman, D., Hexner, E., 2017; Hay, K.A., Turtle, C.J., 2017; Mohanty et al., 2019). Several studies and clinical trials have been and continue to be carried out to develop and improve CAR-T and CAR-NK cell therapies. Previous studies have demonstrated excellent therapeutic efficacy, encouraging the continued development of these therapies, and current studies investigate techniques to improve efficacy, reduce toxicity, and mitigate adverse effects (Littman, D., Hexner, E., 2017; Hay, K.A., Turtle, C.J., 2017; Mohanty et al., 2019).
Além disso, tem sido foco de esforço constante o aprimoramento das técnicas para o desenvolvimento e produção de linfócitos CAR-T, pois as metodologias disponíveis atualmente ainda apresentam dificuldades em relação à qualidade das células, potencial de expansão, persistência, modificações genéticas, entre outros (Fesnak, A. D., 2020; Wang, X., Rivière, I., 2016). Furthermore, the improvement of techniques for the development and production of CAR-T lymphocytes has been the focus of constant effort, as the methodologies currently available still present difficulties in relation to cell quality, expansion potential, persistence, genetic modifications, among others (Fesnak, AD, 2020; Wang, X., Rivière, I., 2016).
Logo, observa-se uma clara necessidade de novas opções de terapias aprimoradas para o tratamento de mieloma múltiplo baseadas em CAR-T e/ou CAR- NK, que sejam mais eficientes e com efeitos adversos amenizados, que visem também superar as dificuldades de desenvolvimento e produção. Therefore, there is a clear need for new, improved therapy options for the treatment of multiple myeloma based on CAR-T and/or CAR-NK, which are more efficient and have reduced adverse effects, and which also aim to overcome development and production difficulties.
Com base no acima exposto, o objetivo da presente invenção é fornecer polinucleotídeos que codificam CARs anti-BCMA com atividade citotóxica para células de mieloma múltiplo, assim como um vetor e uma célula imune efetora que compreendem o mesmo, os quais podem ser usados para o desenvolvimento de imunoterapias de células CAR-T e/ou CAR-NK mais eficientes, buscando amenizar as dificuldades operacionais e/ou os efeitos adversos do produto. Based on the above, the objective of the present invention is to provide polynucleotides encoding anti-BCMA CARs with cytotoxic activity for multiple myeloma cells, as well as a vector and an effector immune cell comprising the same, which can be used for the development of more efficient CAR-T and/or CAR-NK cell immunotherapies, seeking to alleviate the operational difficulties and/or adverse effects of the product.
Assim, os inventores conseguiram surpreendentemente superar os problemas do estado da técnica por meio de polinucleotídeos que codificam receptores de antígeno quimérico (CARs) anti-BCMA, em que os CARs compreendem um domínio de ligação ao antígeno, um domínio transmembrana e um domínio intracelular compreendendo um ou mais domínios coestimulatórios e um ou mais domínios de sinalização, em que o domínio de ligação anti-BCMA compreende o domínio variável de cadeia leve de SEQ ID NO: 1 e o domínio variável de cadeia pesada de SEQ ID NO: 3. Os inventores desenvolveram também um vetor e uma composição, compreendendo uma célula imune efetora, que compreendem o polinucleotídeo, além de um método para a produção de célula imune efetora modificada e do uso do polinucleotídeo, do vetor, da composição ou da célula imune efetora produzida pelo método para a fabricação de um medicamento para o tratamento de mieloma múltiplo. Thus, the inventors have surprisingly managed to overcome the problems of the prior art by means of polynucleotides encoding anti-BCMA chimeric antigen receptors (CARs), in which the CARs comprise an antigen-binding domain, a transmembrane domain and an intracellular domain comprising one or more costimulatory domains and one or more signaling domains, in which the anti-BCMA binding domain comprises the light chain variable domain of SEQ ID NO: 1 and the heavy chain variable domain of SEQ ID NO: 3. The inventors have also developed a vector and a composition, comprising an effector immune cell, comprising the polynucleotide, in addition to a method for the production of a modified effector immune cell and the use of the polynucleotide, the vector, the composition or the effector immune cell produced by the method for the manufacture of a medicament for the treatment of multiple myeloma.
DESCRIÇÃO RESUMIDA DA INVENÇÃO BRIEF DESCRIPTION OF THE INVENTION
A presente invenção está relacionada a um polinucleotídeo que codifica um receptor de antígeno quimérico (CAR) anti-BCMA e a um polipeptídeo que corresponde ao receptor de antígeno quimérico (CAR) anti-BCMA por si. A presente invenção também está relacionada a um vetor e a uma composição, compreendendo uma célula imune efetora, que compreendem o polinucleotídeo. The present invention relates to a polynucleotide encoding an anti-BCMA chimeric antigen receptor (CAR) and to a polypeptide corresponding to the anti-BCMA chimeric antigen receptor (CAR) itself. The present invention also relates to a vector and a composition, comprising an effector immune cell, which comprise the polynucleotide.
A presente invenção também é direcionada a um método para a produção de uma célula imune efetora modificada e ao uso do polinucleotídeo, do vetor, da composição ou da célula imune efetora produzida pelo método para a fabricação de um medicamento para o tratamento de mieloma múltiplo. The present invention is also directed to a method for producing a modified effector immune cell and to the use of the polynucleotide, vector, composition or effector immune cell produced by the method for manufacturing a medicament for the treatment of multiple myeloma.
BREVE DESCRIÇÃO DAS FIGURAS BRIEF DESCRIPTION OF THE FIGURES
As Figuras 1 a, 1 b e 1c mostram os vetores de expressão/transferência do CAR anti-BCMA da presente invenção. Figures 1 a, 1 b and 1c show the anti-BCMA CAR expression/transfer vectors of the present invention.
A Figura 2 mostra o plasmídeo cassete de empacotamento (vetor psPAX2) do vetor da presente invenção. Figure 2 shows the packaging cassette plasmid (psPAX2 vector) of the vector of the present invention.
A Figura 3 mostra o plasmídeo cassete do envelope (vetor pMD2.G) do vetor da presente invenção. Figure 3 shows the envelope cassette plasmid (pMD2.G vector) of the vector of the present invention.
A Figura 4 demonstra a estratégia de gating da imunofenotipagem de linfócitos T. Figure 4 demonstrates the gating strategy for T lymphocyte immunophenotyping.
A Figura 5 mostra os resultados da imunofenotipagem de linfócitos T pré e pós-seleção com beads magnéticas. Figure 5 shows the results of pre- and post-selection T lymphocyte immunophenotyping with magnetic beads.
A Figura 6 equivale a análise representativa da ativação dos linfócitos T após estimulação com Dynabeads CD3/CD28 por 24 horas. Figure 6 is a representative analysis of T lymphocyte activation after stimulation with Dynabeads CD3/CD28 for 24 hours.
A Figura 7 demonstra a estratégia de gating da detecção de linfócitos CAR+ com o kit BCMA CAR Detection. Figure 7 demonstrates the gating strategy for CAR+ lymphocyte detection with the BCMA CAR Detection kit.
A Figura 8 mostra a eficiência de transdução dos CARs anti-BCMA, anti- BCMA(IL-15) e anti-CD19. Figure 8 shows the transduction efficiency of anti-BCMA, anti-BCMA(IL-15), and anti-CD19 CARs.
A Figura 9 retrata a viabilidade e expansão dos linfócitos CAR-T em cultura por 15 dias. Figure 9 depicts the viability and expansion of CAR-T lymphocytes in culture for 15 days.
A Figura 10 corresponde ao desenho experimental do ensaio de calceína. A Figura 11 corresponde às curvas de lise celular obtidas nos ensaios de calceína das células CAR-T e linfócitos não transduzidos co-cultivados com duas linhagens tumorais, A) MM1 S (BCMA+ CD19-) e B) Nalm6 (BCMA-CD19+). Figure 10 corresponds to the experimental design of the calcein assay. Figure 11 corresponds to the cell lysis curves obtained in the calcein assays of CAR-T cells and non-transduced lymphocytes co-cultured with two tumor cell lines, A) MM1 S (BCMA+ CD19-) and B) Nalm6 (BCMA-CD19+).
A Figura 12 demonstra a estratégia de gating para análise ensaio de produção de citocinas. Figure 12 demonstrates the gating strategy for cytokine production assay analysis.
A Figura 13 mostra a produção das citocinas IFN- y (A) e TNF- a (B) pelas populações de linfócitos CAR-T anti-BCMA (com e sem IL-15), linfócitos não- transduzidos e linfócitos CAR-T CD19 em resposta a estímulo por PMA + lonomicina, ou quando co-cultivados com as linhagens tumorais MM1 S e Nalm6. Figure 13 shows the production of the cytokines IFN-γ (A) and TNF-α (B) by populations of anti-BCMA CAR-T lymphocytes (with and without IL-15), non-transduced lymphocytes and CD19 CAR-T lymphocytes in response to stimulation by PMA + lonomycin, or when co-cultured with the MM1 S and Nalm6 tumor lines.
A Figura 14 representa a estratégia de gates utilizada para a imunofenotipagem das células NK pré e pós-seleção. Figure 14 represents the gating strategy used for immunophenotyping of NK cells pre- and post-selection.
A Figura 15 exemplifica a estratégia de gates utilizada para análise das células NK que expressam o CAR anti-BCMA-IL-15. Figure 15 exemplifies the gating strategy used to analyze NK cells expressing the anti-BCMA-IL-15 CAR.
A Figura 16 retrata a eficiência de transdução do CAR anti-BCMA-IL-15 para as células NK e NK memory like (ML). Figure 16 depicts the transduction efficiency of anti-BCMA-IL-15 CAR for NK and memory-like (ML) NK cells.
A Figura 17 mostra a eficiência de transdução do CAR anti-BCMA-IL-15 para as células NK e NK memory like. Figure 17 shows the transduction efficiency of anti-BCMA-IL-15 CAR for NK and memory-like NK cells.
Na Figura 18, pode ser observada a taxa de morte celular para as células NK e NK memory like transduzidas e não transduzidas co-cultivadas com linhagem MM1 S. In Figure 18, the cell death rate for transduced and non-transduced NK and memory-like NK cells co-cultured with the MM1 S cell line can be observed.
A Figura 19 mostra a estratégia de gates utilizada para análise de produção de citocinas das células NK e NK memory like que expressão o CAR anti- BCMA. Figure 19 shows the gating strategy used to analyze cytokine production by NK and memory-like NK cells expressing the anti-BCMA CAR.
A Figura 20 mostra a produção de IFN-y, TNF-a e CD107a por células NK e NK memory like CAR anti-BCMA-IL-15 em resposta ao estímulo por PMA + lonomicina, ou em co-cultura a linhagem tumoral MM1 S. Figure 20 shows the production of IFN-y, TNF-a and CD107a by anti-BCMA-IL-15 NK and NK memory like CAR cells in response to stimulation by PMA + lonomycin, or in co-culture with the MM1 S tumor cell line.
DESCRIÇÃO DETALHADA DA INVENÇÃO DETAILED DESCRIPTION OF THE INVENTION
A presente invenção está relacionada um polinucleotídeo que codifica um receptor de antígeno quimérico (CAR) anti-BCMA, em que o CAR compreende um domínio de ligação anti-BCMA, um domínio transmembrana e um domínio intracelular compreendendo um ou mais domínios coestimulatórios e um domínio de sinalização, em que o domínio de ligação anti-BCMA compreende o domínio variável de cadeia leve de SEQ ID NO: 1 e o domínio variável de cadeia pesada de SEQ ID NO: 3. The present invention relates to a polynucleotide encoding an anti-BCMA chimeric antigen receptor (CAR), wherein the CAR comprises an anti-BCMA binding domain, a transmembrane domain, and an intracellular domain comprising one or more costimulatory domains and a signaling domain, wherein the anti-BCMA binding domain comprises the light chain variable domain of SEQ ID NO: 1 and the heavy chain variable domain of SEQ ID NO: 3.
Os domínios variáveis de cadeia leve e pesada de SEQ ID NOs: 1 e 3 correspondem aos domínios variáveis de cadeia leve e pesada do anticorpo monoclonal de coelho anti-BCMA murino, clone 11 D5.3. Os domínios variáveis de cadeia leve e pesada são codificados pelas sequências de nucleotídeos de SEQ ID Nos: 2 e 4, respectivamente, e as sequências degeneradas das mesmas. The light and heavy chain variable domains of SEQ ID NOs: 1 and 3 correspond to the light and heavy chain variable domains of the rabbit monoclonal anti-murine BCMA antibody, clone 11 D5.3. The light and heavy chain variable domains are encoded by the nucleotide sequences of SEQ ID Nos: 2 and 4, respectively, and degenerate sequences thereof.
Em uma realização preferencial, os domínios variáveis de ligação ao BCMA estão conectados por um ligante peptídico flexível (linker). Em uma realização preferencial, o ligante peptídico é selecionado a partir do grupo que compreende, mas não está limitado a GSTSGSGKPGSGEGSTKG, (G4S)3, (G4S)4, entre outros. Em uma realização preferencial, o ligante peptídico é GSTSGSGKPGSGEGSTKG (SEQ ID NO: 5), codificado pela SEQ ID NO: 6 e pelas sequências degeneradas da mesma. Em outra realização preferencial, o ligante peptídico é (G4S)3 (SEQ ID NO: 7), codificado pela SEQ ID NO: 8 e pelas sequências degeneradas da mesma. In a preferred embodiment, the BCMA-binding variable domains are connected by a flexible peptide linker. In a preferred embodiment, the peptide linker is selected from the group comprising, but not limited to, GSTSGSGKPGSGEGSTKG, (G4S)3, (G4S)4, among others. In a preferred embodiment, the peptide linker is GSTSGSGKPGSGEGSTKG (SEQ ID NO: 5), encoded by SEQ ID NO: 6 and degenerate sequences thereof. In another preferred embodiment, the peptide linker is (G4S)3 (SEQ ID NO: 7), encoded by SEQ ID NO: 8 and degenerate sequences thereof.
Em uma realização preferencial, o domínio de ligação anti-BCMA do CAR codificado é um fragmento variável de cadeia simples (do inglês, Single-Chain Fragment Variable - ScFv), em que os domínios variáveis de cadeia leve e pesada podem estar em qualquer uma das seguintes orientações: domínio variável de cadeia leve-ligante-domínio variável de cadeia pesada ou domínio variável de cadeia pesada- ligante-domínio variável de cadeia leve. Em uma realização preferencial, a orientação do ScFv é domínio variável de cadeia leve-ligante-domínio variável de cadeia pesada. Em outra realização preferencial, a orientação do ScFv é domínio variável de cadeia pesada-ligante-domínio variável de cadeia leve. Em uma realização ainda mais preferencial, o ScFv compreende a sequência de SEQ ID NO: 9, codificada pela SEQ ID NO: 10 e pelas sequências degeneradas da mesma. In a preferred embodiment, the encoded CAR anti-BCMA binding domain is a Single-Chain Fragment Variable (ScFv), wherein the light and heavy chain variable domains can be in either of the following orientations: light chain variable domain-linker-heavy chain variable domain or heavy chain variable domain-linker-light chain variable domain. In a preferred embodiment, the orientation of the ScFv is light chain variable domain-linker-heavy chain variable domain. In another preferred embodiment, the orientation of the ScFv is heavy chain variable domain-linker-light chain variable domain. In an even more preferred embodiment, the ScFv comprises the sequence of SEQ ID NO: 9, encoded by SEQ ID NO: 10. ID NO: 10 and its degenerate sequences.
Em uma realização preferencial, o CAR anti-BCMA codificado inclui um domínio transmembrana que compreende um domínio transmembrana de uma proteína, por exemplo, selecionada do grupo que consiste na cadeia alfa, beta ou zeta do receptor de células T, CD28, CD3 épsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137 e CD 154. Em uma realização mais preferencial, o domínio transmembrana codificado compreende o domínio transmembrana CD8. Em uma realização ainda mais preferencial, o domínio transmembrana codificado compreende o domínio transmembrana CD8 alfa, compreendendo a sequência de SEQ ID NO: 11 , codificada pela SEQ ID NO: 12 e pelas sequências degeneradas da mesma. In a preferred embodiment, the encoded anti-BCMA CAR includes a transmembrane domain comprising a transmembrane domain of a protein, e.g., selected from the group consisting of the T-cell receptor alpha, beta, or zeta chain, CD28, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, and CD 154. In a more preferred embodiment, the encoded transmembrane domain comprises the CD8 transmembrane domain. In an even more preferred embodiment, the encoded transmembrane domain comprises the CD8 alpha transmembrane domain, comprising the sequence of SEQ ID NO: 11, encoded by SEQ ID NO: 12, and degenerate sequences thereof.
Em uma realização preferencial, o domínio de ligação anti-BCMA codificado está conectado ao domínio transmembrana por uma região de dobradiça (hinge). Em uma realização preferencial, a região de dobradiça compreende a região constante de uma molécula IgG 1 , de uma molécula CD8 ou CD28. Em uma realização preferencial, a região de dobradiça compreende CD8 alfa. Em uma realização mais preferencial, a região de dobradiça compreende a sequência de SEQ ID NO: 13, codificada pela SEQ ID NO: 14 e pelas sequências degeneradas da mesma. In a preferred embodiment, the encoded anti-BCMA binding domain is connected to the transmembrane domain by a hinge region. In a preferred embodiment, the hinge region comprises the constant region of an IgG1 molecule, a CD8 or a CD28 molecule. In a preferred embodiment, the hinge region comprises CD8 alpha. In a more preferred embodiment, the hinge region comprises the sequence of SEQ ID NO: 13, encoded by SEQ ID NO: 14 and degenerate sequences thereof.
Em uma realização preferencial, o CAR anti-BCMA codificado inclui peptídeo sinal. Em uma realização preferencial, o peptídeo sinal compreende um peptídeo sinal de cadeia pesada de lgG1 , um peptídeo sinal do receptor 2 do fator estimulador de colônias de granulócitos-macrófagos (GM-CSFR2) ou um peptídeo sinal CD8 alfa. Em uma realização preferencial, o peptídeo sinal compreende um peptídeo sinal CD8 alfa. Em uma realização mais preferencial, o peptídeo sinal compreende a sequência de SEQ ID NO: 15, codificada pela SEQ ID NO: 16 e pelas sequências degeneradas da mesma. In a preferred embodiment, the encoded anti-BCMA CAR includes a signal peptide. In a preferred embodiment, the signal peptide comprises an lgG1 heavy chain signal peptide, a granulocyte-macrophage colony-stimulating factor receptor 2 (GM-CSFR2) signal peptide, or a CD8 alpha signal peptide. In a preferred embodiment, the signal peptide comprises a CD8 alpha signal peptide. In a more preferred embodiment, the signal peptide comprises the sequence of SEQ ID NO: 15, encoded by SEQ ID NO: 16, and degenerate sequences thereof.
Em uma realização preferencial, o CAR anti-BCMA codificado inclui um ou mais domínios coestimulatórios para aumentar a eficácia e expansão das células que expressam os receptores CAR. Tal como aqui utilizado, o termo "domínio coestimulatório" refere-se a um domínio de sinalização intracelular de uma molécula coestimulatória. Moléculas coestimulatórias são moléculas da superfície celular diferentes dos receptores de antígeno ou receptores Fc que fornecem um segundo sinal necessário para ativação e função das células T/NK após a ligação ao antígeno. Em uma realização preferencial, o um ou mais domínios coestimulatórios são selecionados a partir de CD28, CD27, OX-40 (CD134), DAP10, DAP 12, 4-1 BB (CD137), CD40L, 2B4, DNAM, CS1 , CD48, NKG2D, NKp30, NKp44, NKp46, NKp80 e uma combinação dos mesmos. Em uma realização preferencial, o domínio coestimulatório compreende 4-1 BB. Em uma realização mais preferencial, o domínio coestimulatório compreende a sequência de SEQ ID NO: 17, codificada pela SEQ ID NO: 18 e pelas sequências degeneradas da mesma. Em outra realização preferencial, o domínio coestimulatório compreende 2B4. Em uma realização mais preferencial, o domínio coestimulatório compreende a sequência de SEQ ID NO: 19, codificada pela SEQ ID NO: 20 e pelas sequências degeneradas da mesma. In a preferred embodiment, the encoded anti-BCMA CAR includes one or more costimulatory domains to increase cell efficacy and expansion. expressing CAR receptors. As used herein, the term "costimulatory domain" refers to an intracellular signaling domain of a costimulatory molecule. Costimulatory molecules are cell surface molecules other than antigen receptors or Fc receptors that provide a second signal necessary for NK/T cell activation and function following antigen binding. In a preferred embodiment, the one or more costimulatory domains are selected from CD28, CD27, OX-40 (CD134), DAP10, DAP 12, 4-1 BB (CD137), CD40L, 2B4, DNAM, CS1, CD48, NKG2D, NKp30, NKp44, NKp46, NKp80, and a combination thereof. In a preferred embodiment, the costimulatory domain comprises 4-1 BB. In a more preferred embodiment, the costimulatory domain comprises the sequence of SEQ ID NO: 17, encoded by SEQ ID NO: 18 and by degenerate sequences thereof. In another preferred embodiment, the costimulatory domain comprises 2B4. In a more preferred embodiment, the costimulatory domain comprises the sequence of SEQ ID NO: 19, encoded by SEQ ID NO: 20 and by degenerate sequences thereof.
Em uma realização preferencial, o CAR anti-BCMA codificado inclui um domínio de sinalização intracelular que regula a ativação primária do complexo TCR de forma estimulatória ou inibitória. Em realizações preferenciais, o CAR anti-BCMA codificado compreende um domínio de sinalização primário CD3 zeta. Em uma realização mais preferencial, o domínio de sinalização primário CD3 zeta compreende a sequência de SEQ ID NO: 21 , codificada pela SEQ ID NO: 22 e pelas sequências degeneradas da mesma. In a preferred embodiment, the encoded anti-BCMA CAR includes an intracellular signaling domain that regulates primary activation of the TCR complex in a stimulatory or inhibitory manner. In preferred embodiments, the encoded anti-BCMA CAR comprises a CD3 zeta primary signaling domain. In a more preferred embodiment, the CD3 zeta primary signaling domain comprises the sequence of SEQ ID NO: 21, encoded by SEQ ID NO: 22 and degenerate sequences thereof.
Em uma realização preferencial, o CAR anti-BCMA codificado inclui um domínio de sinalização primário CD3 zeta e um ou mais domínios de sinalização coestimulatórios, que podem estar ligados em qualquer ordem ao C-terminal do domínio transmembrana. In a preferred embodiment, the encoded anti-BCMA CAR includes a CD3 zeta primary signaling domain and one or more costimulatory signaling domains, which may be linked in any order to the C-terminus of the transmembrane domain.
Em uma realização preferencial, o CAR anti-BCMA codificado inclui ainda uma citocina. As citocinas são proteínas que regulam a resposta imunológica, incluindo, mas não limitadas as interleucinas e interferon-y (IFN-y). As interleucinas (IL) são produzidas pelos leucócitos em resposta a microrganismos e outros antígenos. Já ao INF-y é atribuída a função de atrair os macrófagos, que auxiliam na remoção de restos celulares e promovem a cicatrização e reorganização das áreas com inflamação. O INF-y é a principal citocina liberada após a indução da resposta imune adaptativa, sendo produzido pelos linfócitos T efetores. Em uma realização mais preferencial, o CAR anti-BCMA codificado inclui interleucina, interferon-y (IFN-y) ou uma combinação dos mesmos. In a preferred embodiment, the encoded anti-BCMA CAR further includes a cytokine. Cytokines are proteins that regulate the immune response, including, but not limited to, interleukins and interferon-y (IFN-y). Interleukins (IL) are produced by leukocytes in response to microorganisms and other antigens. INF-y is assigned the function of attracting macrophages, which assist in the removal of cellular debris and promote healing and reorganization of areas of inflammation. INF-y is the main cytokine released after induction of the adaptive immune response and is produced by effector T lymphocytes. In a more preferred embodiment, the encoded anti-BCMA CAR includes interleukin, interferon-y (IFN-y) or a combination thereof.
Em uma realização mais preferencial, o CAR anti-BCMA codificado inclui uma ou mais interleucinas. Preferencialmente, o CAR anti-BCMA codificado inclui IL- 2, IL-7, IL-15 ou uma combinação das mesmas. A IL-2 é o principal fator de estimulação de células T, como um fator de crescimento e ativação para todas as subpopulações de linfócitos T. Já a IL-7 é uma citocina que estimula o crescimento e maturação de linfócitos B e a ativação de linfócitos T, sendo secretada por células do estroma da medula óssea e do timo. A IL-15 é uma citocina com semelhança estrutural com a IL-2 que se liga por meio de um complexo composto por IL-2/IL-15 da cadeia beta do receptor e a cadeia gama comum. A IL-15 é segregada por fagócitos mononucleares após a infecção por vírus. In a more preferred embodiment, the encoded anti-BCMA CAR includes one or more interleukins. Preferably, the encoded anti-BCMA CAR includes IL-2, IL-7, IL-15 or a combination thereof. IL-2 is the main T-cell stimulating factor, as a growth and activation factor for all T lymphocyte subpopulations. IL-7 is a cytokine that stimulates the growth and maturation of B lymphocytes and the activation of T lymphocytes, and is secreted by bone marrow and thymus stromal cells. IL-15 is a cytokine with structural similarity to IL-2 that binds via a complex composed of IL-2/IL-15 receptor beta chain and common gamma chain. IL-15 is secreted by mononuclear phagocytes after virus infection.
Em uma realização preferencial, o CAR anti-BCMA codificado inclui IL- 15. Em uma realização mais preferencial, a citocina IL-15 compreende a sequência de SEQ ID NO: 23, codificada pela SEQ ID NO: 24 e pelas sequências degeneradas da mesma. Em uma realização mais preferencial, o CAR anti-BCMA codificado inclui IL-2. Em uma realização mais preferencial, a citocina IL-2 compreende a sequência de SEQ ID NO: 25, codificada pela SEQ ID NO: 26 e pelas sequências degeneradas da mesma. In a preferred embodiment, the encoded anti-BCMA CAR includes IL-15. In a more preferred embodiment, the IL-15 cytokine comprises the sequence of SEQ ID NO: 23, encoded by SEQ ID NO: 24 and degenerate sequences thereof. In a more preferred embodiment, the encoded anti-BCMA CAR includes IL-2. In a more preferred embodiment, the IL-2 cytokine comprises the sequence of SEQ ID NO: 25, encoded by SEQ ID NO: 26 and degenerate sequences thereof.
Em uma realização preferencial, o domínio de sinalização do CAR anti- BCMA codificado está conectado à interleucina por meio de uma sequência de divagem. Em uma realização preferencial, a sequência de divagem compreende elementos de sequência 2A ou elementos de sequência semelhantes a 2S, que podem ser usados para a ligação de proteínas ou coexpressão das mesmas. É de conhecimento comum a técnica quais sequências de divagem podem ser utilizadas para coexpressar genes, ligando quadros de leitura abertos para formar um único cístron. In a preferred embodiment, the encoded anti-BCMA CAR signaling domain is connected to interleukin via a cleavage sequence. In a preferred embodiment, the cleavage sequence comprises 2A sequence elements or 2S-like sequence elements, which can be used for protein binding or co-expression. It is common knowledge in the art that cleavage sequences can be used to co-express genes by linking open reading frames to form a single cistron.
Em uma realização preferencial, a sequência de clivagem compreendida no CAR anti-BCMA codificado da presente invenção compreende o vírus da rinite equina (E2A), o vírus da febre aftosa (F2A), o vírus Thosea asigna (T2A), o Teschovírus-1 suíno (P2A) ou uma combinação dos mesmos. Em uma realização preferencial, a sequência de clivagem compreende T2A. Em uma realização mais preferencial, a sequência de clivagem T2A compreende a sequência de SEQ ID NO: 27, codificada pela SEQ ID NO: 28 e pelas sequências degeneradas da mesma. Em uma realização preferencial, a sequência de divagem compreende P2A. Em uma realização mais preferencial, a sequência de divagem P2A compreende a sequência de SEQ ID NO: 29, codificada pela SEQ ID NO: 30 e pelas sequências degeneradas da mesma. Em uma realização preferencial, a sequência de divagem compreende E2A. Em uma realização mais preferencial, a sequência de divagem E2A compreende a sequência de SEQ ID NO: 31 , codificada pela SEQ ID NO: 32 e pelas sequências degeneradas da mesma. In a preferred embodiment, the cleavage sequence comprised in the encoded anti-BCMA CAR of the present invention comprises equine rhinitis virus (E2A), foot-and-mouth disease virus (F2A), Thosea asigna virus (T2A), porcine Teschovirus-1 (P2A), or a combination thereof. In a preferred embodiment, the cleavage sequence comprises T2A. In a more preferred embodiment, the T2A cleavage sequence comprises the sequence of SEQ ID NO: 27, encoded by SEQ ID NO: 28, and degenerate sequences thereof. In a preferred embodiment, the cleavage sequence comprises P2A. In a more preferred embodiment, the P2A cleavage sequence comprises the sequence of SEQ ID NO: 29, encoded by SEQ ID NO: 30, and degenerate sequences thereof. In a preferred embodiment, the cleavage sequence comprises E2A. In a more preferred embodiment, the E2A cleavage sequence comprises the sequence of SEQ ID NO: 31, encoded by SEQ ID NO: 32 and degenerate sequences thereof.
Em uma realização preferencial, o CAR anti-BCMA codificado compreende um peptídeo sinal CD8, um ligante, uma região de dobradiça CD8, um domínio transmembrana CD8, um domínio coestimulatório 4-1 BB e um domínio de sinalização CD3 zeta. In a preferred embodiment, the encoded anti-BCMA CAR comprises a CD8 signal peptide, a linker, a CD8 hinge region, a CD8 transmembrane domain, a 4-1 BB costimulatory domain, and a CD3 zeta signaling domain.
Em uma realização preferencial, o CAR anti-BCMA codificado compreende um peptídeo sinal CD8, um ligante, uma região de dobradiça CD8, um domínio transmembrana CD8, um domínio coestimulatório 4-1 BB, um domínio de sinalização CD3 zeta, uma sequência de divagem T2A e IL-15. In a preferred embodiment, the encoded anti-BCMA CAR comprises a CD8 signal peptide, a linker, a CD8 hinge region, a CD8 transmembrane domain, a 4-1 BB costimulatory domain, a CD3 zeta signaling domain, a T2A cleavage sequence, and IL-15.
Em uma realização preferencial, o CAR anti-BCMA codificado compreende um peptídeo sinal CD8, um ligante, uma região de dobradiça CD8, um domínio transmembrana CD8, um domínio coestimulatório 2B4, um domínio de sinalização CD3 zeta, uma sequência de clivagem T2A e IL-15. In a preferred embodiment, the encoded anti-BCMA CAR comprises a CD8 signal peptide, a linker, a CD8 hinge region, a CD8 transmembrane domain, a 2B4 costimulatory domain, a CD3 zeta signaling domain, a T2A cleavage sequence, and IL-15.
Em uma realização preferencial, o CAR anti-BCMA codificado compreende uma combinação das sequências selecionadas a partir das sequências ímpares das SEQ ID NOs: 1 a 31. In a preferred embodiment, the encoded anti-BCMA CAR comprises a combination of the sequences selected from the odd sequences of SEQ ID NOs: 1 to 31.
Em uma realização preferencial, o CAR anti-BCMA codificado compreende uma das SEQ ID NOs: 33, 35 ou 37. Em uma realização mais preferencial, o CAR anti-BCMA codificado compreende a SEQ ID NO: 33. Em uma realização mais preferencial, o CAR anti-BCMA codificado compreende a SEQ ID NO: 35. Em uma realização mais preferencial, o CAR anti-BCMA codificado compreende a SEQ ID NO: 37. In a preferred embodiment, the encoded anti-BCMA CAR comprises one of SEQ ID NOs: 33, 35 or 37. In a more preferred embodiment, the encoded anti-BCMA CAR comprises SEQ ID NO: 33. In a more preferred embodiment, the encoded anti-BCMA CAR comprises SEQ ID NO: 35. In a more preferred embodiment, the encoded anti-BCMA CAR comprises SEQ ID NO: 37.
Em uma realização preferencial, o polinucleotídeo que codifica o CAR anti-BCMA da presente invenção compreende uma combinação das sequências de nucleotídeos selecionadas a partir das sequências pares das SEQ ID NOs: 2 a 32. In a preferred embodiment, the polynucleotide encoding the anti-BCMA CAR of the present invention comprises a combination of the nucleotide sequences selected from the even sequences of SEQ ID NOs: 2 to 32.
Em uma realização preferencial, o polinucleotídeo que codifica o CAR anti-BCMA da presente invenção compreende uma das SEQ ID NOs: 34, 36 ou 38. Em uma realização mais preferencial, o polinucleotídeo que codifica o CAR anti-BCMA da presente invenção compreende a SEQ ID NO: 34. Em outra realização mais preferencial, o polinucleotídeo que codifica o CAR anti-BCMA da presente invenção compreende a SEQ ID NO: 36. Em outra realização mais preferencial, o polinucleotídeo que codifica o CAR anti-BCMA da presente invenção compreende a SEQ ID NO: 38. In a preferred embodiment, the polynucleotide encoding the anti-BCMA CAR of the present invention comprises one of SEQ ID NOs: 34, 36 or 38. In a more preferred embodiment, the polynucleotide encoding the anti-BCMA CAR of the present invention comprises SEQ ID NO: 34. In another more preferred embodiment, the polynucleotide encoding the anti-BCMA CAR of the present invention comprises SEQ ID NO: 36. In another more preferred embodiment, the polynucleotide encoding the anti-BCMA CAR of the present invention comprises SEQ ID NO: 38.
A presente invenção também se refere a um polipeptídeo que corresponde ao receptor de antígeno quimérico (CAR) que compreende um domínio de ligação anti-BCMA, um domínio transmembrana e um domínio intracelular compreendendo um ou mais domínios coestimulatórios e um domínio de sinalização, em que o domínio de ligação anti-BCMA compreende o domínio variável de cadeia leve de SEQ ID NO: 1 e o domínio variável de cadeia leve de SEQ ID NO: 3. The present invention also relates to a polypeptide corresponding to the chimeric antigen receptor (CAR) comprising an anti-BCMA binding domain, a transmembrane domain and an intracellular domain comprising one or more costimulatory domains and a signaling domain, wherein the anti-BCMA binding domain comprises the β-chain variable domain. light chain variable domain of SEQ ID NO: 1 and the light chain variable domain of SEQ ID NO: 3.
Os domínios variáveis de cadeia leve e pesada de SEQ ID NOs: 1 e 3 do CAR anti-BCMA da presente invenção correspondem aos domínios variáveis de cadeia leve e pesada do anticorpo monoclonal de coelho anti-BCMA murino, clone 11 D5.3. The light and heavy chain variable domains of SEQ ID NOs: 1 and 3 of the anti-BCMA CAR of the present invention correspond to the light and heavy chain variable domains of the rabbit monoclonal anti-murine BCMA antibody, clone 11 D5.3.
Em uma realização preferencial, os domínios variáveis de ligação ao BCMA do CAR da presente invenção estão conectados por um ligante peptídico flexível (linker). Em uma realização preferencial, o ligante peptídico é selecionado a partir de GSTSGSGKPGSGEGSTKG, (G4S)3, (G4S)4, entre outros. Em uma realização preferencial, o ligante peptídico é GSTSGSGKPGSGEGSTKG (SEQ ID NO: 5). Em outra realização preferencial, o ligante peptídico é (G4S)3 (SEQ ID NO: 7). In a preferred embodiment, the BCMA-binding variable domains of the CAR of the present invention are connected by a flexible peptide linker. In a preferred embodiment, the peptide linker is selected from GSTSGSGKPGSGEGSTKG, (G4S)3, (G4S)4, among others. In a preferred embodiment, the peptide linker is GSTSGSGKPGSGEGSTKG (SEQ ID NO: 5). In another preferred embodiment, the peptide linker is (G4S)3 (SEQ ID NO: 7).
Em uma realização preferencial, o domínio de ligação anti-BCMA do CAR da presente invenção é um ScFv, em que os domínios variáveis de cadeia leve e pesada podem estar em qualquer uma das seguintes orientações: domínio variável de cadeia leve-ligante-domínio variável de cadeia pesada ou domínio variável de cadeia pesada-ligante-domínio variável de cadeia leve. Em uma realização preferencial, a orientação do ScFv é domínio variável de cadeia leve-ligante-domínio variável de cadeia pesada. Em outra realização preferencial, a orientação do ScFv é domínio variável de cadeia pesada-ligante-domínio variável de cadeia leve. Em uma realização ainda mais preferencial, o ScFv compreende a sequência de SEQ ID NO: 9. In a preferred embodiment, the anti-BCMA binding domain of the CAR of the present invention is a ScFv, wherein the light and heavy chain variable domains can be in any of the following orientations: light chain variable domain-linker-heavy chain variable domain or heavy chain variable domain-linker-light chain variable domain. In a preferred embodiment, the orientation of the ScFv is light chain variable domain-linker-heavy chain variable domain. In another preferred embodiment, the orientation of the ScFv is heavy chain variable domain-linker-light chain variable domain. In an even more preferred embodiment, the ScFv comprises the sequence of SEQ ID NO: 9.
Em uma realização preferencial, o CAR anti-BCMA da presente invenção inclui um domínio transmembrana que compreende um domínio transmembrana de uma proteína, por exemplo, selecionada a partir da cadeia alfa, beta ou zeta do receptor de células T, CD28, CD3 épsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137 e CD 154. Em uma realização mais preferencial, o domínio transmembrana do CAR anti-BCMA da presente invenção compreende o domínio transmembrane CD8. Em uma realização ainda mais preferencial, o domínio transmembrana do CAR anti-BCMA da presente invenção compreende o domínio transmembrana CD8 alfa, compreendendo a sequência de SEQ ID NO: 11 . In a preferred embodiment, the anti-BCMA CAR of the present invention includes a transmembrane domain comprising a transmembrane domain of a protein, e.g., selected from the alpha, beta or zeta chain of the T cell receptor, CD28, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137 and CD 154. In a more preferred embodiment, the transmembrane domain of ... of a protein, e.g., selected from the alpha, beta or zeta chain of the T cell receptor, CD28, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137 and CD 154. The present invention comprises the CD8 transmembrane domain. In an even more preferred embodiment, the transmembrane domain of the anti-BCMA CAR of the present invention comprises the CD8 alpha transmembrane domain, comprising the sequence of SEQ ID NO: 11.
Em uma realização preferencial, o domínio de ligação anti-BCMA do CAR da presente invenção está conectado ao domínio transmembrana por uma região de dobradiça (hinge). Em uma realização preferencial, a região de dobradiça compreende a região constante de uma molécula lgG1 , de uma molécula CD8 ou CD28. Em uma realização preferencial, a região de dobradiça compreende CD8 alfa. Em uma realização mais preferencial, a região de dobradiça do CAR anti-BCMA da presente invenção compreende a sequência de SEQ ID NO: 13, codificada pela SEQ ID NO: 14. In a preferred embodiment, the anti-BCMA binding domain of the CAR of the present invention is connected to the transmembrane domain by a hinge region. In a preferred embodiment, the hinge region comprises the constant region of an lgG1 molecule, a CD8 or a CD28 molecule. In a preferred embodiment, the hinge region comprises CD8 alpha. In a more preferred embodiment, the hinge region of the anti-BCMA CAR of the present invention comprises the sequence of SEQ ID NO: 13, encoded by SEQ ID NO: 14.
Em uma realização preferencial, o CAR anti-BCMA da presente invenção inclui um peptídeo sinal. Em uma realização preferencial, o peptídeo sinal compreende um peptídeo sinal de cadeia pesada de lgG1 , peptídeo sinal do receptor 2 do fator estimulador de colônias de granulócitos-macrófagos (GM-CSFR2) ou um peptídeo sinal CD8 alfa. Em uma realização preferencial, o peptídeo sinal compreende um peptídeo sinal CD8 alfa. Em uma realização mais preferencial, o peptídeo sinal do CAR anti-BCMA da presente invenção compreende a sequência de SEQ ID NO: 15. In a preferred embodiment, the anti-BCMA CAR of the present invention includes a signal peptide. In a preferred embodiment, the signal peptide comprises an lgG1 heavy chain signal peptide, granulocyte-macrophage colony-stimulating factor receptor 2 (GM-CSFR2) signal peptide, or a CD8 alpha signal peptide. In a preferred embodiment, the signal peptide comprises a CD8 alpha signal peptide. In a more preferred embodiment, the signal peptide of the anti-BCMA CAR of the present invention comprises the sequence of SEQ ID NO: 15.
Em uma realização preferencial, o CAR anti-BCMA da presente invenção inclui um ou mais domínios coestimulatórios para aumentar a eficácia e expansão das células que expressam o mesmo. Em uma realização preferencial, o um ou mais domínios coestimulatórios do CAR anti-BCMA da presente invenção são selecionados de, mas não limitados a CD28, CD27, OX-40 (CD134), DAP10, DAP 12, 4-1 BB (CD137), CD40L, 2B4, DNAM, CS1 , CD48, NKG2D, NKp30, NKp44, NKp46, NKp80 e uma combinação dos mesmos. Em uma realização preferencial, o domínio coestimulatório do CAR anti-BCMA da presente invenção compreende 4-1 BB. Em uma realização mais preferencial, o domínio coestimulatório 4-1 BB compreende a sequência de SEQ ID NO: 17. Em outra realização preferencial, o domínio coestimulatório do CAR anti-BCMA da presente invenção compreende 2B4. Em uma realização mais preferencial, o domínio coestimulatório 2B4 compreende a sequência de SEQ ID NO: 19. In a preferred embodiment, the anti-BCMA CAR of the present invention includes one or more costimulatory domains to increase the efficacy and expansion of cells expressing the same. In a preferred embodiment, the one or more costimulatory domains of the anti-BCMA CAR of the present invention are selected from, but not limited to, CD28, CD27, OX-40 (CD134), DAP10, DAP 12, 4-1 BB (CD137), CD40L, 2B4, DNAM, CS1, CD48, NKG2D, NKp30, NKp44, NKp46, NKp80, and a combination thereof. In a preferred embodiment, the costimulatory domain of the anti-BCMA CAR of the present invention comprises 4-1 BB. In a more preferred embodiment, the 4-1 BB costimulatory domain comprises sequence of SEQ ID NO: 17. In another preferred embodiment, the costimulatory domain of the anti-BCMA CAR of the present invention comprises 2B4. In a more preferred embodiment, the costimulatory domain 2B4 comprises the sequence of SEQ ID NO: 19.
Em uma realização preferencial, o CAR anti-BCMA da presente invenção inclui um domínio de sinalização intracelular. Em realizações preferenciais, o CAR anti-BCMA da presente invenção compreende um domínio de sinalização primário CD3 zeta. Em uma realização mais preferencial, o domínio de sinalização primário CD3 zeta compreende a sequência de SEQ ID NO: 21 . In a preferred embodiment, the anti-BCMA CAR of the present invention includes an intracellular signaling domain. In preferred embodiments, the anti-BCMA CAR of the present invention comprises a CD3 zeta primary signaling domain. In a more preferred embodiment, the CD3 zeta primary signaling domain comprises the sequence of SEQ ID NO: 21.
Em uma realização preferencial, o CAR anti-BCMA da presente invenção inclui um domínio de sinalização primário CD3 zeta e um ou mais domínios de sinalização coestimulatórios, que podem estar ligados em qualquer ordem ao C- terminal do domínio transmembrana. In a preferred embodiment, the anti-BCMA CAR of the present invention includes a CD3 zeta primary signaling domain and one or more costimulatory signaling domains, which may be linked in any order to the C-terminus of the transmembrane domain.
Em uma realização preferencial, o CAR anti-BCMA da presente invenção inclui ainda uma citocina. Em uma realização mais preferencial, o CAR anti- BCMA da presente invenção inclui interleucina, interferon-y (IFN-y) ou uma combinação dos mesmos. In a preferred embodiment, the anti-BCMA CAR of the present invention further includes a cytokine. In a more preferred embodiment, the anti-BCMA CAR of the present invention includes interleukin, interferon-γ (IFN-γ), or a combination thereof.
Em uma realização mais preferencial, o CAR anti-BCMA da presente invenção inclui uma ou mais interleucinas. Preferencialmente, o CAR da presente invenção inclui IL-2, IL-7, IL-15 ou uma combinação das mesmas. In a more preferred embodiment, the anti-BCMA CAR of the present invention includes one or more interleukins. Preferably, the CAR of the present invention includes IL-2, IL-7, IL-15 or a combination thereof.
Em uma realização preferencial, o CAR anti-BCMA da presente invenção inclui IL-15. Em uma realização mais preferencial, a citocina IL-15 compreende a sequência de SEQ ID NO: 23. Em uma realização preferencial, o CAR anti-BCMA inclui IL-2. Em uma realização mais preferencial, a citocina IL-2 compreende a sequência de SEQ ID NO: 25. In a preferred embodiment, the anti-BCMA CAR of the present invention includes IL-15. In a more preferred embodiment, the IL-15 cytokine comprises the sequence of SEQ ID NO: 23. In a preferred embodiment, the anti-BCMA CAR includes IL-2. In a more preferred embodiment, the IL-2 cytokine comprises the sequence of SEQ ID NO: 25.
Em uma realização preferencial, o domínio de sinalização está conectado à citocina por meio de uma sequência de divagem. Em uma realização preferencial, a sequência de divagem compreende elementos de sequência 2A ou elementos de sequência semelhantes a 2S. In a preferred embodiment, the signaling domain is connected to the cytokine via a cleavage sequence. In a preferred embodiment, the cleavage sequence comprises sequence elements 2A or 2S-like sequence elements.
Em uma realização preferencial, o CAR anti-BCMA da presente invenção compreende a sequência de divagem que compreende o vírus da rinite equina (E2A), o vírus da febre aftosa (F2A), o vírus Thosea asigna (T2A), o Teschovírus-1 suíno (P2A) ou uma combinação dos mesmos. Em uma realização preferencial, a sequência de clivagem compreende T2A. Em uma realização mais preferencial, a sequência de clivagem T2A compreende a sequência de SEQ ID NO: 27. Em uma realização preferencial, a sequência de divagem compreende P2A. Em uma realização mais preferencial, a sequência de divagem P2A compreende a sequência de SEQ ID NO: 29. Em uma realização preferencial, a sequência de divagem compreende E2A. Em uma realização mais preferencial, a sequência de divagem E2A compreende a sequência de SEQ ID NO: 31 . In a preferred embodiment, the anti-BCMA CAR of the present invention comprises the cleavage sequence comprising equine rhinitis virus (E2A), foot-and-mouth disease virus (F2A), Thosea asigna virus (T2A), porcine Teschovirus-1 (P2A) or a combination thereof. In a preferred embodiment, the cleavage sequence comprises T2A. In a more preferred embodiment, the T2A cleavage sequence comprises the sequence of SEQ ID NO: 27. In a preferred embodiment, the cleavage sequence comprises P2A. In a more preferred embodiment, the P2A cleavage sequence comprises the sequence of SEQ ID NO: 29. In a preferred embodiment, the cleavage sequence comprises E2A. In a more preferred embodiment, the E2A cleavage sequence comprises the sequence of SEQ ID NO: 31.
Em uma realização preferencial, o CAR anti-BCMA da presente invenção compreende um peptídeo sinal CD8, um ligante, uma região de dobradiça CD8, um domínio transmembrana CD8, um domínio coestimulatório 4-1 BB e um domínio de sinalização CD3 zeta. In a preferred embodiment, the anti-BCMA CAR of the present invention comprises a CD8 signal peptide, a linker, a CD8 hinge region, a CD8 transmembrane domain, a 4-1 BB costimulatory domain, and a CD3 zeta signaling domain.
Em uma realização preferencial, o CAR anti-BCMA da presente invenção compreende um peptídeo sinal CD8, um ligante, uma região de dobradiça CD8, um domínio transmembrana CD8, um domínio coestimulatório 4-1 BB, um domínio de sinalização CD3 zeta, uma sequência de divagem T2A e IL-15. In a preferred embodiment, the anti-BCMA CAR of the present invention comprises a CD8 signal peptide, a linker, a CD8 hinge region, a CD8 transmembrane domain, a 4-1 BB costimulatory domain, a CD3 zeta signaling domain, a T2A cleavage sequence, and IL-15.
Em uma realização preferencial, o CAR anti-BCMA da presente invenção compreende um peptídeo sinal CD8, um ligante, uma região de dobradiça CD8, um domínio transmembrana CD8, um domínio coestimulatório 2B4, um domínio de sinalização CD3 zeta, uma sequência de divagem T2A e IL-15. In a preferred embodiment, the anti-BCMA CAR of the present invention comprises a CD8 signal peptide, a linker, a CD8 hinge region, a CD8 transmembrane domain, a 2B4 costimulatory domain, a CD3 zeta signaling domain, a T2A cleavage sequence, and IL-15.
Em uma realização preferencial, o CAR anti-BCMA da presente invenção compreende uma combinação das sequências selecionadas a partir das sequências ímpares das SEQ ID NOs: 1 a 31. In a preferred embodiment, the anti-BCMA CAR of the present invention comprises a combination of the sequences selected from the odd sequences of SEQ ID NOs: 1 to 31.
Em uma realização preferencial, o CAR anti-BCMA da presente invenção compreende uma das SEQ ID NOs: 33, 35 ou 37. Em uma realização mais preferencial, o CAR anti-BCMA da presente invenção compreende a SEQ ID NO: 33. Em uma realização mais preferencial, o CAR anti-BCMA da presente invenção compreende a SEQ ID NO: 35. Em uma realização mais preferencial, o CAR anti- BCMA da presente invenção compreende a SEQ ID NO: 37. In a preferred embodiment, the anti-BCMA CAR of the present invention comprises one of SEQ ID NOs: 33, 35 or 37. In a more preferred embodiment, the anti-BCMA CAR of the present invention comprises SEQ ID NO: 33. In a more preferred embodiment, the anti-BCMA CAR of the present invention comprises SEQ ID NO: 35. In a more preferred embodiment, the anti-BCMA CAR of the present invention comprises SEQ ID NO: 37.
A presente invenção também se refere a um vetor que compreende o polinucleotídeo que codifica um receptor de antígeno quimérico (CAR) anti-BCMA, em que o CAR compreende um domínio de ligação anti-BCMA, um domínio transmembrana e um domínio intracelular compreendendo um ou mais domínios coestimulatórios e um domínio de sinalização, em que o domínio de ligação anti- BCMA compreende o domínio variável de cadeia leve de SEQ ID NO: 1 e o domínio variável de cadeia pesada de SEQ ID NO: 3. The present invention also relates to a vector comprising the polynucleotide encoding an anti-BCMA chimeric antigen receptor (CAR), wherein the CAR comprises an anti-BCMA binding domain, a transmembrane domain, and an intracellular domain comprising one or more costimulatory domains and a signaling domain, wherein the anti-BCMA binding domain comprises the light chain variable domain of SEQ ID NO: 1 and the heavy chain variable domain of SEQ ID NO: 3.
Em uma realização preferencial, o vetor compreende o polinucleotídeo que codifica o CAR anti-BCMA da presente invenção, conforme aqui definido. In a preferred embodiment, the vector comprises the polynucleotide encoding the anti-BCMA CAR of the present invention, as defined herein.
Em uma realização mais preferencial, o vetor compreende o polinucleotídeo que codifica o CAR anti-BCMA compreendendo uma das SEQ ID NOs: 34, 36 ou 38. Em uma realização mais preferencial, o polinucleotídeo que codifica o CAR anti-BCMA da presente invenção compreende a SEQ ID NO: 34. Em outra realização mais preferencial, o polinucleotídeo que codifica o CAR anti-BCMA da presente invenção compreende a SEQ ID NO: 36. Em outra realização mais preferencial, o polinucleotídeo que codifica o CAR anti-BCMA da presente invenção compreende a SEQ ID NO: 38. In a more preferred embodiment, the vector comprises the polynucleotide encoding the anti-BCMA CAR comprising one of SEQ ID NOs: 34, 36 or 38. In a more preferred embodiment, the polynucleotide encoding the anti-BCMA CAR of the present invention comprises SEQ ID NO: 34. In another more preferred embodiment, the polynucleotide encoding the anti-BCMA CAR of the present invention comprises SEQ ID NO: 36. In another more preferred embodiment, the polynucleotide encoding the anti-BCMA CAR of the present invention comprises SEQ ID NO: 38.
Em uma realização preferencial, o vetor da presente invenção é um plasm ideo ou um vetor de expressão ou de transferência. In a preferred embodiment, the vector of the present invention is a plasmid or an expression or transfer vector.
Em uma realização preferencial, o vetor da presente invenção compreende um ou mais promotores. Em uma realização preferencial, o um ou mais promotores são selecionados a partir de promotor de citomegalovírus (CMV), um promotor do vírus do sarcoma de Rous (RSV) ou um promotor do vírus símio 40 (SV40). Em uma realização preferencial, o vetor da presente invenção compreende um ou mais terminadores. In a preferred embodiment, the vector of the present invention comprises one or more promoters. In a preferred embodiment, the one or more promoters are selected from a cytomegalovirus (CMV) promoter, a Rous sarcoma virus (RSV) promoter or a simian virus promoter. (SV40). In a preferred embodiment, the vector of the present invention comprises one or more terminators.
Em uma realização preferencial, o vetor da presente invenção compreende um ou mais sítios de reconhecimento de enzimas de restrição. In a preferred embodiment, the vector of the present invention comprises one or more restriction enzyme recognition sites.
Em uma realização preferencial, o vetor da presente invenção compreende um ou mais elementos regulatórios. Os elementos regulatórios podem compreender, sem estarem limitados a origens de replicação, marcadores de resistência a antibióticos, sinais de poliadenilação, elementos regulatórios pós- transcricionais como o do vírus da hepatite B (HPRE) ou do vírus da hepatite de marmota (WPRE), entre outros. In a preferred embodiment, the vector of the present invention comprises one or more regulatory elements. The regulatory elements may comprise, but are not limited to, origins of replication, antibiotic resistance markers, polyadenylation signals, post-transcriptional regulatory elements such as those of the hepatitis B virus (HPRE) or the woodchuck hepatitis virus (WPRE), among others.
Em uma realização preferencial, o vetor da presente invenção é um vetor virai. Em uma realização preferencial, o vetor da presente invenção é um vetor virai selecionado a partir de, mas não limitado a retrovirus, adenovirus e vírus adenoassociado. Em uma realização preferencial, o vetor da presente invenção é um adenovirus. Em outra realização preferencial, o vetor da presente invenção é um vírus adenoassociado. Em outra realização preferencial, o vetor da presente invenção é um retrovirus. Em uma realização mais preferencial, o vetor da presente invenção é um retrovirus selecionado a partir de, mas não limitado a um vetor lentiviral e um vetor gamma-retroviral. In a preferred embodiment, the vector of the present invention is a viral vector. In a preferred embodiment, the vector of the present invention is a viral vector selected from, but not limited to, retroviruses, adenoviruses, and adeno-associated viruses. In a preferred embodiment, the vector of the present invention is an adenovirus. In another preferred embodiment, the vector of the present invention is an adeno-associated virus. In another preferred embodiment, the vector of the present invention is a retrovirus. In a more preferred embodiment, the vector of the present invention is a retrovirus selected from, but not limited to, a lentiviral vector and a gamma-retroviral vector.
Em uma realização mais preferencial, vetor da presente invenção é um vetor gamma-retroviral. Em outra realização mais preferencial, o vetor da presente invenção é um vetor lentiviral. Em uma realização preferencial, o vetor lentiviral da presente invenção é selecionado a partir de, mas não limitado ao vírus da imunodeficiência humana (HIV) 1 , vírus da imunodeficiência humana (HIV) 2, vírus da imunodeficiência símia (SIV), vírus da imunodeficiência bovina (BIV), vírus da imunodeficiência felina (FIV) etc. In a more preferred embodiment, the vector of the present invention is a gamma-retroviral vector. In another more preferred embodiment, the vector of the present invention is a lentiviral vector. In a preferred embodiment, the lentiviral vector of the present invention is selected from, but not limited to, human immunodeficiency virus (HIV) 1, human immunodeficiency virus (HIV) 2, simian immunodeficiency virus (SIV), bovine immunodeficiency virus (BIV), feline immunodeficiency virus (FIV), etc.
Em uma realização preferencial, o vetor da presente invenção compreende ainda genes de organização do vírus. Em uma realização preferencial, o vetor compreende um ou mais genes que codificam proteínas estruturais, enzimas necessárias para a replicação, proteínas do envelope virai, proteínas acessórias, proteínas regulatórias e combinações dos mesmos. In a preferred embodiment, the vector of the present invention further comprises virus organization genes. In a preferred embodiment, the vector comprises one or more genes encoding structural proteins, enzymes required for replication, viral envelope proteins, accessory proteins, regulatory proteins, and combinations thereof.
Em uma realização preferencial, o vetor da presente invenção é um HIV compreendendo os genes de organização do vírus, denominados gag, pol e env. Em uma realização preferencial, o vetor da presente invenção codifica ainda proteínas acessórias, como por exemplo, mas não limitadas a Nef, Vpr, Vif e Vpu e proteínas regulatórias, como por exemplo, mas não limitadas a tat e rev. Em uma realização preferencial, o vetor da presente invenção possui sequências específicas necessárias para a sinalização de processos como exportação para o núcleo, sinalizado pelo RRE, integração no genoma e expressão, sinalizados pelas Long Terminal Repeats (LTRs), empacotamento do RNA nos novos virions formados, sinalizados pelo sinal de empacotamento i , e um trato central de polipurina (cPPT). In a preferred embodiment, the vector of the present invention is an HIV comprising the virus organization genes, called gag, pol and env. In a preferred embodiment, the vector of the present invention further encodes accessory proteins, such as, but not limited to, Nef, Vpr, Vif and Vpu and regulatory proteins, such as, but not limited to, tat and rev. In a preferred embodiment, the vector of the present invention has specific sequences necessary for signaling processes such as export to the nucleus, signaled by the RRE, integration into the genome and expression, signaled by the Long Terminal Repeats (LTRs), packaging of the RNA into the newly formed virions, signaled by the packaging signal i, and a central polypurine tract (cPPT).
Em uma realização preferencial, o vetor da presente invenção compreende também a sequência Kozak (gccacc). In a preferred embodiment, the vector of the present invention also comprises the Kozak sequence (gccacc).
Em uma realização preferencial, os genes dos componentes do HIV estão presentes em um único plasm ideo. Em uma realização preferencial, os genes dos componentes do HIV estão presentes em plasmídeos separados. In a preferred embodiment, the genes for the HIV components are present on a single plasmid. In a preferred embodiment, the genes for the HIV components are present on separate plasmids.
Em uma realização preferencial, o vetor da presente invenção corresponde a um vetor lentiviral de primeira geração em que os componentes do HIV estão separados em três plasmídeos. Em outra realização preferencial, o vetor da presente invenção corresponde a um vetor lentiviral de segunda geração em que os componentes do HIV estão separados em três plasmídeos e os genes acessórios vif, vpr, vpu, e nef foram removidos. Em outra realização preferencial, o vetor da presente invenção corresponde a um vetor lentiviral de terceira geração, em que os componentes do HIV estão separados em quatro plasmídeos, os genes vif, vpr, vpu, nef e tat foram removidos e a porção LTR 5’ do plasm ideo de transferência foi substituído por um promotor forte. Em uma realização preferencial, o vetor da presente invenção é um vetor lentiviral de segunda geração que compreende: In a preferred embodiment, the vector of the present invention corresponds to a first generation lentiviral vector in which the HIV components are separated into three plasmids. In another preferred embodiment, the vector of the present invention corresponds to a second generation lentiviral vector in which the HIV components are separated into three plasmids and the accessory genes vif, vpr, vpu, and nef have been removed. In another preferred embodiment, the vector of the present invention corresponds to a third generation lentiviral vector in which the HIV components are separated into four plasmids, the vif, vpr, vpu, nef, and tat genes have been removed, and the 5' LTR portion of the transfer plasmid has been replaced by a strong promoter. In a preferred embodiment, the vector of the present invention is a second generation lentiviral vector comprising:
- um vetor de expressão/transferência; - an expression/transfer vector;
- um plasmídeo cassete de empacotamento; e - a packaging cassette plasmid; and
- um plasmídeo cassete de envelope. - an envelope cassette plasmid.
Em uma realização preferencial, o vetor de expressão/trânsferência do CAR anti-BCMA da presente invenção compreende um vetor de primeira, segunda, terceira ou quarta geração. Preferencialmente, o vetor de expressão do CAR anti- BCMA da presente invenção compreende um vetor de segunda ou quarta geração. Em uma realização mais preferencial, o vetor de expressão do CAR anti-BCMA da presente invenção é selecionado de, mas não limitado ao plasmídeo p4BC, plasmídeo pB4CI, entre outros conhecidos da técnica, que podem ser desenhados e sintetizados de acordo com práticas comuns ao técnico no assunto. Em uma realização preferencial, o vetor de expressão do CAR anti-BCMA da presente invenção compreende o polinucleotídeo que codifica os domínios variáveis de cadeia leve e pesada do clone 11 D5.3. In a preferred embodiment, the anti-BCMA CAR expression/transfer vector of the present invention comprises a first, second, third or fourth generation vector. Preferably, the anti-BCMA CAR expression vector of the present invention comprises a second or fourth generation vector. In a more preferred embodiment, the anti-BCMA CAR expression vector of the present invention is selected from, but not limited to, plasmid p4BC, plasmid pB4CI, among others known in the art, which can be designed and synthesized according to practices common to the person skilled in the art. In a preferred embodiment, the anti-BCMA CAR expression vector of the present invention comprises the polynucleotide encoding the light and heavy chain variable domains of clone 11 D5.3.
Em uma realização preferencial, o vetor de expressão do CAR anti- BCMA da presente invenção compreende o polinucleotídeo conforme aqui descrito. Em uma realização preferencial, o vetor de expressão do CAR anti-BCMA da presente invenção compreende as sequências de nucleotídeos de SEQ ID Nos: 2 e 4. Em uma realização preferencial, o vetor de expressão do CAR anti-BCMA da presente invenção compreende uma combinação de duas ou mais sequências de nucleotídeos selecionadas a partir das sequências pares das SEQ ID NOs: 2 a 32. Em uma realização mais preferencial, o vetor de expressão do CAR anti-BCMA da presente invenção compreende qualquer uma das SEQ ID NOs: 34, 36 ou 38. Em uma realização mais preferencial, o vetor de expressão do CAR anti-BCMA da presente invenção compreende ainda um ou mais elementos comuns a vetores de expressão como um ou mais promotores, uma ou mais origens de replicação, um ou mais terminadores, um ou mais marcadores de antibiótico, um ou mais sítios de reconhecimento de enzimas de restrição, um ou mais sinais de poliadenilação, entre outros. Em uma realização mais preferencial, o vetor de expressão do CAR anti-BCMA da presente invenção compreende ainda um ou mais elementos comuns a vetores de expressão de SEQ ID Nos: 39 a 53. In a preferred embodiment, the anti-BCMA CAR expression vector of the present invention comprises the polynucleotide as described herein. In a preferred embodiment, the anti-BCMA CAR expression vector of the present invention comprises the nucleotide sequences of SEQ ID NOs: 2 and 4. In a preferred embodiment, the anti-BCMA CAR expression vector of the present invention comprises a combination of two or more nucleotide sequences selected from the even-numbered sequences of SEQ ID NOs: 2 to 32. In a more preferred embodiment, the anti-BCMA CAR expression vector of the present invention comprises any one of SEQ ID NOs: 34, 36 or 38. In a more preferred embodiment, the anti-BCMA CAR expression vector of the present invention further comprises one or more elements common to expression vectors such as one or more promoters, one or more origins of replication, one or more terminators, one or more antibiotic markers, one or more restriction enzyme recognition sites, one or more polyadenylation signals, among others. In a more preferred embodiment, the anti-BCMA CAR expression vector of the present invention further comprises one or more elements common to expression vectors of SEQ ID Nos: 39 to 53.
Em uma realização mais preferencial, o vetor de expressão do CAR anti- BCMA da presente invenção compreende qualquer um dos vetores conforme ilustrados nas Figuras 1 a, 1 b e 1 c. In a more preferred embodiment, the anti-BCMA CAR expression vector of the present invention comprises any of the vectors as illustrated in Figures 1 a, 1 b and 1 c.
Em uma realização preferencial, o plasmídeo cassete de empacotamento da presente invenção é, mas não limitado ao plasmídeo psPAX2 (Addgene, #12260), que contém os genes lentivirais gag, pol e RRE. O vetor psPAX2 é retratado na Figura 2. In a preferred embodiment, the packaging cassette plasmid of the present invention is, but is not limited to, the psPAX2 plasmid (Addgene, #12260), which contains the lentiviral gag, pol and RRE genes. The psPAX2 vector is depicted in Figure 2.
Em uma realização preferencial, o plasmídeo cassete do envelope é, mas não limitado ao plasmídeo pMD2.G (Addgene, #12259), que contém as sequências codificadoras do envelope pseudotipado do vírus VSV-G. O vetor pMD2.G é retratado na Figura 3. In a preferred embodiment, the envelope cassette plasmid is, but is not limited to, plasmid pMD2.G (Addgene, #12259), which contains the envelope coding sequences pseudotyped to the VSV-G virus. The pMD2.G vector is depicted in Figure 3.
A presente invenção também se refere a uma composição compreendendo uma célula imune efetora que compreende o polinucleotídeo ou o vetor da presente invenção, conforme aqui descritos. The present invention also relates to a composition comprising an effector immune cell comprising the polynucleotide or vector of the present invention as described herein.
Em uma realização preferencial, a composição da presente invenção é uma composição farmacêutica. A expressão “composição farmacêutica” designa uma preparação que se encontra em uma forma que permite que a atividade biológica de um ingrediente ativo nela contido seja efetiva. In a preferred embodiment, the composition of the present invention is a pharmaceutical composition. The term “pharmaceutical composition” designates a preparation that is in a form that allows the biological activity of an active ingredient contained therein to be effective.
Em uma realização preferencial, a composição da presente invenção compreende uma célula imune efetora compreendendo o polinucleotídeo que codifica um receptor de antígeno quimérico (CAR) anti-BCMA, em que o CAR compreende um domínio de ligação anti-BCMA, um domínio transmembrana e um domínio intracelular compreendendo um ou mais domínios coestimulatórios e um domínio de sinalização, em que o domínio de ligação anti-BCMA compreende o domínio variável de cadeia leve de SEQ ID NO: 1 e o domínio variável de cadeia pesada de SEQ ID NO: 3, codificados pelas sequências de nucleotídeos de SEQ ID Nos: 2 e 4, respectivamente, e as sequências degeneradas das mesmas. In a preferred embodiment, the composition of the present invention comprises an effector immune cell comprising the polynucleotide encoding an anti-BCMA chimeric antigen receptor (CAR), wherein the CAR comprises an anti-BCMA binding domain, a transmembrane domain and an intracellular domain comprising one or more costimulatory domains and a signaling domain, wherein the anti-BCMA binding domain comprises the light chain variable domain of SEQ ID NO: 1 and the heavy chain variable domain of SEQ ID NO: 3, encoded by the nucleotide sequences of SEQ ID Nos: 2 and 4, respectively, and degenerate sequences thereof.
Em outra realização preferencial, a composição da presente invenção compreende uma célula imune efetora compreendendo o vetor. Em uma realização mais preferencial, o vetor compreende um vetor de expressão compreendendo o polinucleotídeo que codifica o CAR anti-BCMA compreendendo uma das SEQ ID NOs: 34, 36 ou 38. Em uma realização mais preferencial, o polinucleotídeo que codifica o CAR anti-BCMA da presente invenção compreende a SEQ ID NO: 34. Em outra realização mais preferencial, o polinucleotídeo que codifica o CAR anti-BCMA da presente invenção compreende a SEQ ID NO: 36. Em outra realização mais preferencial, o polinucleotídeo que codifica o CAR anti-BCMA da presente invenção compreende a SEQ ID NO: 38. Em uma realização preferencial, a célula imune efetora é transduzida com o vetor da presente invenção. In another preferred embodiment, the composition of the present invention comprises an effector immune cell comprising the vector. In a more preferred embodiment, the vector comprises an expression vector comprising the polynucleotide encoding the anti-BCMA CAR comprising one of SEQ ID NOs: 34, 36 or 38. In a more preferred embodiment, the polynucleotide encoding the anti-BCMA CAR of the present invention comprises SEQ ID NO: 34. In another more preferred embodiment, the polynucleotide encoding the anti-BCMA CAR of the present invention comprises SEQ ID NO: 36. In another more preferred embodiment, the polynucleotide encoding the anti-BCMA CAR of the present invention comprises SEQ ID NO: 38. In a preferred embodiment, the effector immune cell is transduced with the vector of the present invention.
Em uma realização preferencial, a célula imune efetora compreende, mas não está limitada a linfócitos T, células NK ou uma combinação das mesmas. Em uma realização preferencial, a célula imune efetora compreende linfócitos T. Em uma realização preferencial, a célula imune efetora compreende células NK. Em uma realização preferencial, a célula imune efetora compreende linfócitos T e células NK. In a preferred embodiment, the effector immune cell comprises, but is not limited to, T lymphocytes, NK cells, or a combination thereof. In a preferred embodiment, the effector immune cell comprises T lymphocytes. In a preferred embodiment, the effector immune cell comprises NK cells. In a preferred embodiment, the effector immune cell comprises both T lymphocytes and NK cells.
Em uma realização preferencial, a célula imune efetora é produzida pelo método da presente invenção. In a preferred embodiment, the effector immune cell is produced by the method of the present invention.
Em uma realização preferencial, a composição da presente invenção compreende ainda um ou mais excipientes, carreadores ou diluentes. Em uma realização preferencial, os excipientes, carreadores ou diluentes são farmaceuticamente aceitáveis conhecidos da técnica, incluindo, mas não limitados a adjuvantes, carreadores, excipientes, deslizantes, agentes edulcorantes, diluentes, conservantes, corantes, intensificadores de sabor, tensoativos, agentes umectantes, agentes dispersantes, agentes de suspensão, estabilizantes, solventes, emulsificantes aceitáveis para uso em humanos ou animais. Preferencialmente, o um ou mais excipientes, carreadores ou diluentes compreendem açúcares, amidos, celulose e seus derivados, gelatina, talco, manteiga de cacau, ceras, gorduras animais e vegetais, parafinas, silicones, bentonitas, ácido silícico, óxido de zinco, óleos, glicóis, polióis, ésteres, ágar; agentes tamponantes, ácido algínico, água, solução salina, solução de Ringer, álcoois, soluções tampão de fosfato, e quaisquer outras substâncias compatíveis utilizadas em composições farmacêuticas. Os profissionais da área são altamente capazes para determinar os excipientes, carreadores e diluentes farmaceuticamente aceitáveis para cada composição e finalidade. In a preferred embodiment, the composition of the present invention further comprises one or more excipients, carriers or diluents. In a preferred embodiment, the excipients, carriers or diluents are pharmaceutically acceptable excipients known in the art, including, but not limited to, adjuvants, carriers, excipients, glidants, sweetening agents, diluents, preservatives, colorants, flavor enhancers, surfactants, wetting agents, dispersing agents, suspending agents, stabilizers, solvents, emulsifiers acceptable for use in humans or animals. Preferably, the one or more excipients, carriers or diluents comprise sugars, starches, cellulose and its derivatives, gelatin, talc, cocoa butter, waxes, animal and vegetable fats, paraffins, silicones, bentonites, silicic acid, zinc oxide, oils, glycols, polyols, esters, agar; buffering agents, alginic acid, water, saline, Ringer's solution, alcohols, phosphate buffer solutions, and any other compatible substances used in pharmaceutical compositions. Those skilled in the art are highly capable of determining the pharmaceutically acceptable excipients, carriers and diluents for each composition and purpose.
Em uma realização preferencial, a composição da presente invenção compreende uma quantidade de células efetoras imunes que expressam CAR contempladas neste documento. Tal como aqui utilizado, o termo "quantidade" refere- se a "uma quantidade eficaz" ou "uma quantidade terapeuticamente eficaz" de uma célula geneticamente modificada, por exemplo, célula T ou NK, para alcançar um resultado profilático ou terapêutico. Os profissionais da área são altamente capazes para determinar a quantidade terapeuticamente eficaz para cada composição e finalidade. In a preferred embodiment, the composition of the present invention comprises an amount of CAR-expressing immune effector cells contemplated herein. As used herein, the term "amount" refers to "an effective amount" or "a therapeutically effective amount" of a genetically modified cell, e.g., T or NK cell, to achieve a prophylactic or therapeutic result. Those skilled in the art are highly skilled in determining the therapeutically effective amount for each composition and purpose.
Em uma realização preferencial, a composição da presente invenção pode compreender agentes adicionais, como por exemplo, mas sem caráter limitativo, citocinas, fatores de crescimento, hormônios, quimioterapêuticos, radioterapêuticos, pró-fármacos, fármacos, anticorpos etc. In a preferred embodiment, the composition of the present invention may comprise additional agents, such as, but not limited to, cytokines, growth factors, hormones, chemotherapeutics, radiotherapeutics, prodrugs, drugs, antibodies, etc.
Adicionalmente, a presente invenção está relacionada a um método para produção de célula imune efetora modificada, compreendendo a introdução na célula imune efetora do polinucleotídeo ou do vetor da presente invenção, conforme aqui descritos. Additionally, the present invention relates to a method for producing a modified effector immune cell, comprising introducing into the effector immune cell the polynucleotide or vector of the present invention, as described herein.
Em uma realização preferencial, o método da presente invenção compreende a introdução na célula imune efetora do polinucleotídeo que codifica um receptor de antígeno quimérico (CAR) anti-BCMA, em que o CAR compreende urn domínio de ligação anti-BCMA, um domínio transmembrana e um domínio intracelular compreendendo um ou mais domínios coestimulatórios e um domínio de sinalização, em que o domínio de ligação anti-BCMA compreende o domínio variável de cadeia leve de SEQ ID NO: 1 e o domínio variável de cadeia pesada de SEQ ID NO: 3, codificados pelas sequências de nucleotídeos de SEQ ID Nos: 2 e 4, respectivamente, e as sequências degeneradas das mesmas. In a preferred embodiment, the method of the present invention comprises introducing into the effector immune cell the polynucleotide encoding a anti-BCMA chimeric antigen receptor (CAR), wherein the CAR comprises an anti-BCMA binding domain, a transmembrane domain, and an intracellular domain comprising one or more costimulatory domains and a signaling domain, wherein the anti-BCMA binding domain comprises the light chain variable domain of SEQ ID NO: 1 and the heavy chain variable domain of SEQ ID NO: 3, encoded by the nucleotide sequences of SEQ ID Nos: 2 and 4, respectively, and degenerate sequences thereof.
Em outra realização preferencial, o método da presente invenção compreende a introdução na célula imune efetora do vetor. Em uma realização mais preferencial, o vetor compreende um vetor de expressão compreendendo o polinucleotídeo que codifica o CAR anti-BCMA compreendendo uma das SEQ ID NOs: 34, 36 ou 38. Em uma realização mais preferencial, o polinucleotídeo que codifica o CAR anti-BCMA da presente invenção compreende a SEQ ID NO: 34. Em outra realização mais preferencial, o polinucleotídeo que codifica o CAR anti-BCMA da presente invenção compreende a SEQ ID NO: 36. Em outra realização mais preferencial, o polinucleotídeo que codifica o CAR anti-BCMA da presente invenção compreende a SEQ ID NO: 38. In another preferred embodiment, the method of the present invention comprises introducing into the effector immune cell the vector. In a more preferred embodiment, the vector comprises an expression vector comprising the polynucleotide encoding the anti-BCMA CAR comprising one of SEQ ID NOs: 34, 36 or 38. In a more preferred embodiment, the polynucleotide encoding the anti-BCMA CAR of the present invention comprises SEQ ID NO: 34. In another more preferred embodiment, the polynucleotide encoding the anti-BCMA CAR of the present invention comprises SEQ ID NO: 36. In another more preferred embodiment, the polynucleotide encoding the anti-BCMA CAR of the present invention comprises SEQ ID NO: 38.
Em uma realização preferencial, a célula imune efetora compreende, mas não está limitada a linfócitos T, células NK ou uma combinação das mesmas. Em uma realização preferencial, a célula imune efetora compreende linfócitos T. Em uma realização preferencial, a célula imune efetora compreende células NK. Em uma realização preferencial, a célula imune efetora compreende linfócitos T e células NK. In a preferred embodiment, the effector immune cell comprises, but is not limited to, T lymphocytes, NK cells, or a combination thereof. In a preferred embodiment, the effector immune cell comprises T lymphocytes. In a preferred embodiment, the effector immune cell comprises NK cells. In a preferred embodiment, the effector immune cell comprises both T lymphocytes and NK cells.
Em uma realização preferencial, a célula imune efetora é transduzida com o vetor da presente invenção. Em uma realização preferencial, o vetor da presente invenção é um vetor lentiviral. In a preferred embodiment, the effector immune cell is transduced with the vector of the present invention. In a preferred embodiment, the vector of the present invention is a lentiviral vector.
Em uma realização preferencial, o método da presente invenção compreende, antes da introdução do polinucleotídeo ou do vetor da presente invenção na célula imune efetora, a transfecção transiente do vetor de expressão e dos plasm ídeos cassetes de empacotamento e do envelope para a produção das partículas virais. In a preferred embodiment, the method of the present invention comprises, prior to the introduction of the polynucleotide or vector of the present invention into the effector immune cell, the transient transfection of the expression vector and the plasmids packaging and envelope cassettes for the production of viral particles.
Em uma realização preferencial, a transfecção transiente é feita com células HEK (do inglês, Human Embryonic Kidney”). Particularmente, as células HEK de empacotamento compreendem HEK293T. In a preferred embodiment, transient transfection is done with HEK (Human Embryonic Kidney) cells. In particular, the packaging HEK cells comprise HEK293T.
Em uma realização preferencial, o vetor de expressão e os plasm ídeos cassetes de empacotamento e do envelope são misturados e gotejados sobre as células HEK293T, que produzirão as partículas virais compreendendo os polinucleotídeos da presente invenção, em que as partículas virais produzidas correspondem ao vetor da presente invenção. In a preferred embodiment, the expression vector and the packaging and envelope cassette plasmids are mixed and dripped onto HEK293T cells, which will produce viral particles comprising the polynucleotides of the present invention, wherein the viral particles produced correspond to the vector of the present invention.
Em uma realização preferencial, as partículas virais serão utilizadas para a transdução da célula imune efetora de interesse. In a preferred embodiment, the viral particles will be used for transduction of the effector immune cell of interest.
Em uma realização preferencial, a célula imune efetora utilizada no método da presente invenção compreende, mas não está limitada a linfócitos T, células NK ou uma combinação das mesmas. Em uma realização preferencial, a célula imune efetora compreende linfócitos T. Em uma realização preferencial, a célula imune efetora compreende células NK. Em uma realização preferencial, a célula imune efetora compreende linfócitos T e células NK. In a preferred embodiment, the effector immune cell used in the method of the present invention comprises, but is not limited to, T lymphocytes, NK cells, or a combination thereof. In a preferred embodiment, the effector immune cell comprises T lymphocytes. In a preferred embodiment, the effector immune cell comprises NK cells. In a preferred embodiment, the effector immune cell comprises both T lymphocytes and NK cells.
Em uma realização preferencial, a fonte das células imune efetoras utilizadas no método da presente invenção é selecionada a partir de, mas não limitada ao sangue periférico ou cordão umbilical. In a preferred embodiment, the source of the immune effector cells used in the method of the present invention is selected from, but not limited to, peripheral blood or umbilical cord blood.
Em uma realização preferencial, o método da presente invenção compreende, antes da introdução do polinucleotídeo ou do vetor da presente invenção na célula imune efetora, as etapas de coleta, isolamento, seleção e ativação das células imune efetoras. In a preferred embodiment, the method of the present invention comprises, prior to the introduction of the polynucleotide or vector of the present invention into the effector immune cell, the steps of collecting, isolating, selecting and activating the effector immune cells.
Em uma realização preferencial, o isolamento das células imune efetoras é realizado por gradiente de concentração. Demais técnicas para o isolamento das células imune efetoras de interesse são conhecidas da técnica e podem ser usadas de forma alternativa às descritas na presente invenção. In a preferred embodiment, the isolation of the immune effector cells is performed by concentration gradient. Other techniques for isolating the immune effector cells of interest are known in the art and can be used in an alternative way to those described in the present invention.
Em uma realização preferencial, a seleção das células imune efetoras é realizada através de seleção magnética por meio de kits disponíveis comercialmente ou beads. In a preferred embodiment, selection of immune effector cells is performed through magnetic selection using commercially available kits or beads.
Em uma realização preferencial, a ativação das células imune efetoras é realizada por meio de Dynabeads CD3/CD28 ou interleucinas adequadas para cada tipo de célula, por exemplo, como as interleucinas 12, 15 e 18. In a preferred embodiment, activation of immune effector cells is performed by means of Dynabeads CD3/CD28 or interleukins appropriate for each cell type, for example, such as interleukins 12, 15 and 18.
Em uma realização preferencial, o método da presente invenção compreende ainda uma etapa de identificação das células imune efetoras, que pode ser realizada antes e/ou depois da introdução do polinucleotídeo ou do vetor. Em uma realização preferencial, a identificação das células imune efetoras é realizada por imunofenotipagem. Demais técnicas para a identificação das células imune efetoras são conhecidas e podem ser usadas de forma alternativa à descrita na presente invenção. In a preferred embodiment, the method of the present invention further comprises a step of identifying the immune effector cells, which can be performed before and/or after the introduction of the polynucleotide or the vector. In a preferred embodiment, the identification of the immune effector cells is performed by immunophenotyping. Other techniques for identifying the immune effector cells are known and can be used in an alternative manner to that described in the present invention.
Em uma realização preferencial, a etapa de introdução do polinucleotídeo ou do vetor nas células imune efetoras do método da presente invenção é feita por meio de transdução. In a preferred embodiment, the step of introducing the polynucleotide or vector into the immune effector cells of the method of the present invention is carried out by means of transduction.
Em uma realização preferencial, a transdução compreende a adição das partículas virais produzidas às células imunes efetoras. Em uma realização preferencial, após a transdução, células feeder podem ser adicionadas a cultura das células imune efetoras modificadas. In a preferred embodiment, transduction comprises the addition of the produced viral particles to the effector immune cells. In a preferred embodiment, following transduction, feeder cells can be added to the culture of the modified effector immune cells.
Em uma realização preferencial, o método da presente invenção compreende ainda a expansão das células imune modificadas. In a preferred embodiment, the method of the present invention further comprises expanding the modified immune cells.
Em uma realização preferencial, o método da presente invenção compreende ainda etapas de avaliação da eficiência da transdução, da viabilidade, da expansão, da citotoxicidade e da produção de citocinas das células imune efetoras modificadas. In a preferred embodiment, the method of the present invention further comprises steps for evaluating the transduction efficiency, viability, expansion, cytotoxicity and cytokine production of the modified immune effector cells.
Em uma realização preferencial, a avaliação da eficiência da transdução é realizada por meio de kits comerciais que detectam CARs direcionados ao antígeno BCMA, seguidas de citometria de fluxo. Demais técnicas são amplamente conhecidas e podem ser utilizadas de forma alternativa. In a preferred embodiment, the assessment of transduction efficiency is performed using commercial kits that detect CARs directed to the BCMA antigen, followed by flow cytometry. Other techniques are widely known and can be used as alternatives.
Em uma realização preferencial, a avaliação da citotoxicidade é realizada por meio do ensaio com calceína acetoximetil (AM), seguido de citometria de fluxo. Demais técnicas são amplamente conhecidas e podem ser utilizadas de forma alternativa. In a preferred embodiment, cytotoxicity assessment is performed using the acetoxymethyl calcein (AM) assay, followed by flow cytometry. Other techniques are widely known and can be used alternatively.
Em uma realização preferencial, a avaliação da produção de citocinas é realizada por meio da estimulação das células com linhagens BCMA positivas e negativas, seguida de citometria de fluxo, permitindo também a avaliação da especificidade das células imune efetoras modificadas. In a preferred embodiment, the evaluation of cytokine production is performed by stimulating cells with BCMA positive and negative lines, followed by flow cytometry, also allowing the evaluation of the specificity of the modified immune effector cells.
De forma adicional, a presente invenção está relacionada ao uso do polinucleotídeo, do receptor de antígeno quimérico, do vetor, da composição ou da célula imune efetora produzida pelo método da presente invenção, conforme aqui descritos, para a fabricação de um medicamento para o tratamento de mieloma múltiplo. Additionally, the present invention relates to the use of the polynucleotide, chimeric antigen receptor, vector, composition or effector immune cell produced by the method of the present invention, as described herein, for the manufacture of a medicament for the treatment of multiple myeloma.
DEFINIÇÕES DEFINITIONS
Salvo indicação em contrário, os termos usados no presente pedido devem ser entendidos de acordo com uso convencional pelos técnicos no assunto. Unless otherwise indicated, the terms used in this application should be understood in accordance with conventional usage by those skilled in the art.
O termo “célula mononuclear do sangue periférico” (PBMC) como aqui utilizado se refere às células com um núcleo redondo, consistindo em linfócitos (células T, células B, células Natural Killer (NK)) e monócitos, enquanto os eritrócitos e as plaquetas não têm núcleo, e os granulócitos (neutrófilos, basófilos e eosinófilos) têm núcleos multilobados. The term “peripheral blood mononuclear cell” (PBMC) as used herein refers to cells with a round nucleus, consisting of lymphocytes (T cells, B cells, natural killer (NK) cells) and monocytes, whereas erythrocytes and platelets lack a nucleus, and granulocytes (neutrophils, basophils, and eosinophils) have multilobed nuclei.
Os termos “linfócito T” e “célula T” como aqui utilizados correspondem aos componentes do sistema imune adaptativo cuja principal função é a defesa contra agentes danosos. Essas células são derivadas de células tronco hematopoiéticas presentes na medula óssea e passam pelo processo de maturação no timo, onde são selecionados os linfócitos que reagem contra antígenos a eles apresentados (Famili et al., 2017). The terms “T lymphocyte” and “T cell” as used herein correspond to components of the adaptive immune system whose main function is defense against harmful agents. These cells are derived from hematopoietic stem cells present in the bone marrow and undergo the maturation process in the thymus, where they are lymphocytes that react against antigens presented to them are selected (Famili et al., 2017).
Uma característica que identifica os linfócitos T é a expressão do receptor de células T (TCR, do inglês, T Cell Receptor), que consiste em um heterodímero transmembrana composto de duas cadeias polipeptídicas covalentemente ligadas entre si (em sua maioria, a e [3). A função do TCR é reconhecer antígenos apresentados e, posteriormente, ativar o linfócito T para que este inicie suas funções efetoras. A transdução do sinal de ativação é feita por moléculas acessórias fisicamente ligadas ao TCR, sendo elas as proteínas CD3 e (Mariuzza et al. , 2020). A characteristic that identifies T lymphocytes is the expression of the T cell receptor (TCR), which consists of a transmembrane heterodimer composed of two polypeptide chains covalently linked to each other (mostly a and [3]. The function of the TCR is to recognize presented antigens and, subsequently, activate the T lymphocyte so that it begins its effector functions. The transduction of the activation signal is done by accessory molecules physically linked to the TCR, these being the proteins CD3 and (Mariuzza et al., 2020).
O termo “célula NK” como aqui utilizado se refere às células exterminadoras naturais ou células NK (do inglês, Natural Killer), um tipo de linfócitos necessário para o funcionamento do sistema imunológico inato pela sua atividade citotóxica sem a necessidade de reconhecimento prévio de um antígeno específico, contrariamente ao funcionamento dos linfócitos T. The term “NK cell” as used here refers to natural killer cells or NK cells, a type of lymphocyte necessary for the functioning of the innate immune system due to its cytotoxic activity without the need for prior recognition of a specific antigen, contrary to the functioning of T lymphocytes.
As células NK podem ser obtidas de fontes autólogas ou alogênicas que incluem sangue periférico, medula óssea, células-tronco embrionárias humanas, células-tronco pluripotentes induzidas, cordão umbilical ou linhagens celulares NK facilmente disponíveis como NK92. Em uma realização preferencial, as células NK usadas na presente invenção são obtidas do sangue periférico. Em outra realização preferencial, as células NK usadas na presente invenção são provenientes de cordão umbilical (CB). Particularmente, o cordão umbilical é uma fonte alogênica prontamente disponível, de fácil obtenção, e que não requer a submissão de doadores saudáveis aos riscos da coleta. As frequências de células NK no CB é de ~ 15-20%. Para superar o pequeno volume de sangue em uma unidade de CB, tem-se cultivado células NK com “feeder cell” irradiadas, ou seja, juntamente com células apresentadoras de antígenos artificiais (aAPCs) K562 manufaturadas expressando ligantes como IL-21 e 4-1 BB na membrana, proporcionando a proliferação e ativação de células NK de forma confiável e com doses suficientes para imunoterapia adotiva (Shah et al., 2013).NK cells can be obtained from autologous or allogeneic sources that include peripheral blood, bone marrow, human embryonic stem cells, induced pluripotent stem cells, umbilical cord blood or readily available NK cell lines such as NK92. In a preferred embodiment, the NK cells used in the present invention are obtained from peripheral blood. In another preferred embodiment, the NK cells used in the present invention are derived from umbilical cord (CB). In particular, umbilical cord blood is a readily available allogeneic source that is easily obtained and does not require healthy donors to undergo the risks of collection. The frequencies of NK cells in CB are ~15-20%. To overcome the small blood volume in a CB unit, NK cells have been cultured with irradiated feeder cells, i.e., together with manufactured K562 artificial antigen presenting cells (aAPCs) expressing ligands such as IL-21 and 4-1 BB on the membrane, providing proliferation and activation of NK cells in a reliable and with sufficient doses for adoptive immunotherapy (Shah et al., 2013).
O termo “cerca de” aqui utilizado refere-se a um valor de 5 a 10% para mais ou para menos dos valores aos quais se refere. The term “about” used here refers to a value of 5 to 10% more or less than the values to which it refers.
O termo “viabilidade celular” conforme aqui utilizado se refere à análise das células metabolicamente ativas em uma cultura celular e/ou amostra, a fim de avaliar sua atividade qualitativa e/ou quantitativamente. Diversas técnicas para determinar a viabilidade celular são conhecidas da técnica e podem ser utilizadas de forma alternativa ou concomitante. The term “cell viability” as used herein refers to the analysis of metabolically active cells in a cell culture and/or sample in order to assess their activity qualitatively and/or quantitatively. Several techniques for determining cell viability are known in the art and may be used alternatively or concomitantly.
Conforme aqui utilizado, o termo “expansão de linfócitos” corresponde à expansão clonal de linfócitos. A expansão clonal se inicia pela ativação dos linfócitos através da apresentação do antígeno de interesse por meio das moléculas do complexo de histocompatibilidade de classe II (MHC II) das APCs para os receptores dos linfócitos T (TCRs). Essa apresentação promovida pela APC é chamada de “estímulo”. O reconhecimento do antígeno permite a interação dos complexos de moléculas coestimuladoras e a secreção de citocinas, que por sua vez, garantem a sobrevivência de células T específicas para o antígeno e ativam fatores que promovem a exposição de segmentos gênicos específicos levando a diferenciação celular dos linfócitos subdivididos em subtipos (Th1 , Th2 e Th17), que podem ser identificados de acordo com a citocina que mais produzem (INF-y, IL-4 e IL-21 , respectivamente). As used herein, the term “lymphocyte expansion” corresponds to the clonal expansion of lymphocytes. Clonal expansion begins with the activation of lymphocytes through the presentation of the antigen of interest by means of the major histocompatibility complex (MHC II) molecules of APCs to T lymphocyte receptors (TCRs). This presentation promoted by APCs is called “stimulation”. Antigen recognition allows the interaction of costimulatory molecule complexes and the secretion of cytokines, which in turn ensure the survival of antigen-specific T cells and activate factors that promote the exposure of specific gene segments, leading to the cellular differentiation of lymphocytes subdivided into subtypes (Th1, Th2 and Th17), which can be identified according to the cytokine they produce the most (INF-γ, IL-4 and IL-21, respectively).
Conforme aqui utilizado, o termo “sequências degeneradas” corresponde às sequências de nucleotídeos contendo bases degeneradas, ou seja, codificam o mesmo polipeptídeo devido à redundância do código genético. As used here, the term “degenerate sequences” corresponds to nucleotide sequences containing degenerate bases, that is, they encode the same polypeptide due to the redundancy of the genetic code.
Conforme aqui utilizado, o termo “célula imune efetora” corresponde às células do sistema imunológico que respondem a suas células alvo sem a necessidade de coestimulação, sendo normalmente recrutadas da circulação, ativadas por antígenos apresentados por macrófagos e secretam citocinas. As used herein, the term “effector immune cell” corresponds to cells of the immune system that respond to their target cells without the need for co-stimulation, being normally recruited from the circulation, activated by antigens presented by macrophages and secreting cytokines.
O termo “medicamento” conforme aqui utilizado se refere a um produto adequado para terapia/tratamento de mieloma múltiplo que pode compreender, mas não está limitado ao polinucleotídeo que codifica o CAR anti-BCMA da presente invenção, ao vetor da presente invenção, ao CAR da presente invenção, à composição da presente invenção e à célula imune efetora modificada produzida pelo método da presente invenção. The term “drug” as used herein refers to a product suitable for therapy/treatment of multiple myeloma which may comprise, but is not limited to, the polynucleotide encoding the anti-BCMA CAR of the present invention, the vector of the present invention, the CAR of the present invention, the composition of the present invention and the modified effector immune cell produced by the method of the present invention.
Em uma realização preferencial, o medicamento compreende ainda um ou mais excipientes, carreadores ou diluentes. Em uma realização preferencial, os excipientes, carreadores ou diluentes são farmaceuticamente aceitáveis conhecidos da técnica, incluindo, mas não limitados a adjuvantes, carreadores, excipientes, deslizantes, agentes edulcorantes, diluentes, conservantes, corantes, intensificadores de sabor, tensoativos, agentes umectantes, agentes dispersantes, agentes de suspensão, estabilizantes, solventes, emulsificantes aceitáveis para uso em humanos ou animais. Preferencialmente, o um ou mais excipientes, carreadores ou diluentes compreendem açúcares, amidos, celulose e seus derivados, gelatina, talco, manteiga de cacau, ceras, gorduras animais e vegetais, parafinas, silicones, bentonitas, ácido silícico, óxido de zinco, óleos, glicóis, polióis, ésteres, ágar; agentes tamponantes, ácido algínico, água, solução salina, solução de Ringer, álcoois, soluções tampão de fosfato, e quaisquer outras substâncias compatíveis utilizadas em composições farmacêuticas. Os profissionais da área são altamente capazes para determinar os excipientes, carreadores e diluentes farmaceuticamente aceitáveis para cada medicamento e finalidade. In a preferred embodiment, the medicament further comprises one or more excipients, carriers or diluents. In a preferred embodiment, the excipients, carriers or diluents are pharmaceutically acceptable ones known in the art, including, but not limited to, adjuvants, carriers, excipients, glidants, sweetening agents, diluents, preservatives, colorants, flavor enhancers, surfactants, wetting agents, dispersing agents, suspending agents, stabilizers, solvents, emulsifiers acceptable for use in humans or animals. Preferably, the one or more excipients, carriers or diluents comprise sugars, starches, cellulose and its derivatives, gelatin, talc, cocoa butter, waxes, animal and vegetable fats, paraffins, silicones, bentonites, silicic acid, zinc oxide, oils, glycols, polyols, esters, agar; buffering agents, alginic acid, water, saline, Ringer's solution, alcohols, phosphate buffer solutions, and any other compatible substances used in pharmaceutical compositions. Professionals in the field are highly capable of determining the pharmaceutically acceptable excipients, carriers, and diluents for each drug and purpose.
Em uma realização preferencial, o medicamento compreende uma quantidade terapeuticamente eficaz do polinucleotídeo, do receptor de antígeno quimérico, do vetor, da célula imune efetora produzida pelo método ou da composição. Os profissionais da área são altamente capazes para determinar a quantidade terapeuticamente eficaz do polinucleotídeo, do receptor de antígeno quimérico, do vetor, da célula imune efetora produzida pelo método ou da composição para alcançar um resultado profilático ou terapêutico para cada medicamento e finalidade. Em uma realização preferencial, o medicamento pode compreender um ou mais agentes adicionais, como por exemplo, mas sem caráter limitativo, citocinas, fatores de crescimento, hormônios, quimioterapêuticos, radioterapêuticos, pró- fármacos, fármacos, anticorpos etc. In a preferred embodiment, the medicament comprises a therapeutically effective amount of the polynucleotide, chimeric antigen receptor, vector, method-produced immune effector cell, or composition. Those skilled in the art are highly skilled in determining the therapeutically effective amount of the polynucleotide, chimeric antigen receptor, vector, method-produced immune effector cell, or composition to achieve a prophylactic or therapeutic result for each medicament and intended use. In a preferred embodiment, the medicament may comprise one or more additional agents, such as, but not limited to, cytokines, growth factors, hormones, chemotherapeutics, radiotherapeutics, prodrugs, drugs, antibodies, etc.
Em uma realização preferencial, o polinucleotídeo, o receptor de antígeno quimérico, o vetor, a célula imune efetora produzida pelo método ou a composição e o agente adicional estão contidos em uma única formulação ou em formulações diferentes. Em uma realização preferencial, o polinucleotídeo, o receptor de antígeno quimérico, o vetor, a célula imune efetora produzida pelo método ou a composição e o um ou mais agentes adicionais estão contidos em uma única formulação. Em uma realização preferencial, o polinucleotídeo, o receptor de antígeno quimérico, o vetor, a célula imune efetora produzida pelo método ou a composição e o um ou mais agentes adicionais estão contidos em formulações diferentes. In a preferred embodiment, the polynucleotide, the chimeric antigen receptor, the vector, the effector immune cell produced by the method or composition, and the additional agent are contained in a single formulation or in different formulations. In a preferred embodiment, the polynucleotide, the chimeric antigen receptor, the vector, the effector immune cell produced by the method or composition, and the one or more additional agents are contained in a single formulation. In a preferred embodiment, the polynucleotide, the chimeric antigen receptor, the vector, the effector immune cell produced by the method or composition, and the one or more additional agents are contained in different formulations.
Em uma realização preferencial, o polinucleotídeo, o receptor de antígeno quimérico, o vetor, a célula imune efetora produzida pelo método ou a composição e o um ou mais agentes adicionais podem ser administrados juntos ou sequencialmente. In a preferred embodiment, the polynucleotide, the chimeric antigen receptor, the vector, the effector immune cell produced by the method or composition, and the one or more additional agents can be administered together or sequentially.
Adicionalmente, a presente invenção está relacionada a um método para produção de célula imune efetora modificada, compreendendo a introdução na célula imune efetora do polinucleotídeo ou do vetor da presente invenção, conforme aqui descritos. Additionally, the present invention relates to a method for producing a modified effector immune cell, comprising introducing into the effector immune cell the polynucleotide or vector of the present invention, as described herein.
Em uma realização preferencial, o medicamento pode compreender um ou mais agentes terapêuticos adicionais In a preferred embodiment, the medicament may comprise one or more additional therapeutic agents.
O termo “transdução” conforme aqui utilizado se refere ao processo de transferência de DNA ou RNA por meio de um vírus, inspirado no processo de reprodução bacteriano, no qual o DNA é transferido de uma bactéria para outra por meio de bacteriófagos. Preferencialmente, a plataforma para transferência gênica para introdução do transgene CAR da presente invenção nas células imune efetoras é a transdução mediada por vírus, também chamado de vetor virai de expressão ou transferência. Demais plataformas para transferência gênica são conhecidas da técnica e podem ser utilizadas de forma alternativa a técnica aqui descrita, como por exemplo, mas não limitada a transposons Sleeping Beauty, transposons piggyBac, ou transfecção de RNA mensageiro. The term “transduction” as used herein refers to the process of transferring DNA or RNA by means of a virus, inspired by the bacterial reproduction process, in which DNA is transferred from one bacterium to another by means of bacteriophages. Preferably, the gene transfer platform for introducing the CAR transgene of the present invention into the immune effector cells is viral-mediated transduction, also called viral expression or transfer vector. Other platforms for gene transfer are known in the art and can be used as alternatives to the technique described here, such as, but not limited to, Sleeping Beauty transposons, piggyBac transposons, or messenger RNA transfection.
Conforme aqui descrito, o vetor virai adequado para a transdução do transgene CAR da presente invenção nas células imune efetoras é selecionado a partir de, mas não limitado a retrovirus, adenovirus e vírus adenoassociado. Em uma realização preferencial, o vetor da presente invenção é um adenovirus. Em outra realização preferencial, o vetor da presente invenção é um vírus adenoassociado. Em outra realização preferencial, o vetor da presente invenção é um retrovirus. Em uma realização mais preferencial, o vetor da presente invenção é um retrovirus selecionado a partir de, mas não limitado a um vetor lentiviral e um vetor gamma-retroviral. As described herein, the viral vector suitable for transducing the CAR transgene of the present invention into immune effector cells is selected from, but not limited to, retroviruses, adenoviruses, and adeno-associated viruses. In a preferred embodiment, the vector of the present invention is an adenovirus. In another preferred embodiment, the vector of the present invention is an adeno-associated virus. In another preferred embodiment, the vector of the present invention is a retrovirus. In a more preferred embodiment, the vector of the present invention is a retrovirus selected from, but not limited to, a lentiviral vector and a gamma-retroviral vector.
Em uma realização mais preferencial, vetor da presente invenção é um vetor gamma-retroviral. Em outra realização mais preferencial, o vetor da presente invenção é um vetor lentiviral. Em uma realização preferencial, o vetor lentiviral da presente invenção é selecionado a partir de, mas não limitado ao vírus da imunodeficiência humana (HIV). Demais vetores virais são conhecidos da técnica e podem ser utilizados de forma alternativa a técnica aqui descrita. In a more preferred embodiment, the vector of the present invention is a gamma-retroviral vector. In another more preferred embodiment, the vector of the present invention is a lentiviral vector. In a preferred embodiment, the lentiviral vector of the present invention is selected from, but not limited to, the human immunodeficiency virus (HIV). Other viral vectors are known in the art and can be used as alternatives to the technique described herein.
O termo “transfecção” conforme aqui utilizado corresponde ao processo de introdução de ácidos nucleicos em células eucarióticas. As células podem ser transfectadas de modo estável para a integração do DNA em seu genoma ou de modo transitório/transiente para expressão proteica com duração temporária. The term “transfection” as used herein refers to the process of introducing nucleic acids into eukaryotic cells. Cells can be transfected in a stable manner for the integration of DNA into their genome or in a transient manner for protein expression of temporary duration.
O termo “confluência máxima” conforme aqui utilizado corresponde à porcentagem máxima da área do meio de crescimento coberta por células aderentes. Para a presente invenção, a confluência máxima é de cerca de 80% a cerca de 100%, preferencialmente cerca de 85%. The term “maximum confluence” as used herein corresponds to the maximum percentage of the area of the growth medium covered by adherent cells. For the present invention, the maximum confluence is about 80% to about 100%, preferably about 85%.
O termo “imunofenotipagem” conforme aqui utilizado se refere a técnica a laser que avalia características celulares. De forma preferencial, a imunofenotipagem é realizada por citometria de fluxo. Demais técnicas de imunofenotipagem celular são conhecidas da técnica e podem ser utilizadas de forma alternativa a técnica aqui descrita. The term “immunophenotyping” as used herein refers to the technique laser that evaluates cellular characteristics. Preferably, immunophenotyping is performed by flow cytometry. Other cellular immunophenotyping techniques are known in the art and can be used as an alternative to the technique described here.
A citometria de fluxo é uma tecnologia usada para detectar e medir aspectos físicos e químicos de partículas. A estratégia de “gating” corresponde ao princípio básico para a análise da diversidade de informações fornecidas pelas hipóteses testadas na citometria de fluxo, por meio do refinamento e da identificação sequencial das populações de células em pesquisa. Flow cytometry is a technology used to detect and measure physical and chemical aspects of particles. The “gating” strategy corresponds to the basic principle for analyzing the diversity of information provided by the hypotheses tested in flow cytometry, through the refinement and sequential identification of the cell populations under investigation.
EXEMPLOS EXAMPLES
Os Exemplos a seguir não têm a intenção de limitar o escopo das reivindicações da invenção, mas especialmente têm a intenção de ser exemplares de certas realizações. Quaisquer variações que ocorram a um técnico no assunto têm a intenção de ser incluídas no escopo da presente invenção. The following Examples are not intended to limit the scope of the claims of the invention, but rather are intended to be exemplary of certain embodiments. Any variations that occur to one skilled in the art are intended to be included within the scope of the present invention.
EXEMPLO 1 - TRANSFECÇÃO TRANSIENTE DE CÉLULAS EMPACOTADORAS HEK 293T PARA A PRODUÇÃO LENTIVIRAL EXAMPLE 1 - TRANSIENT TRANSFECTION OF HEK 293T PACKAGING CELLS FOR LENTIVIRAL PRODUCTION
As células HEK-293T (ATCC, CRL-3216) foram cultivadas em garrafas T75 (Corning, #43064111) com meio DMEM/F12 (Gibco, #11320-033) suplementado com 10% Soro Fetal Bovino Heat-lnactivated (SFBi) (Gibco, #16140-071 ), 1X L- glutamina (Gibco, #25030-081 ) e 1 % penicilina/estreptomicina (Gibco, 15070-063), até atingirem uma confluência máxima de 85% em menos de dez passagens. Havendo uma quantidade adequada de células, estas foram desaderidas com tripsina (Gibco, #25200-056) , contadas e 1 ,2 x 107 células foram plaqueadas em placa de Petri (Corning, #430167) com 10 ml_ de meio DMEM/F12 suplementado para que a confluência fosse de 95-99% no dia da realização do experimento. HEK-293T cells (ATCC, CRL-3216) were grown in T75 bottles (Corning, #43064111) with DMEM/F12 medium (Gibco, #11320-033) supplemented with 10% Heat-inactivated Fetal Bovine Serum (HFBS) (Gibco, #16140-071), 1X L-glutamine (Gibco, #25030-081), and 1% penicillin/streptomycin (Gibco, 15070-063) until they reached a maximum confluence of 85% in less than ten passages. If there was an adequate amount of cells, they were detached with trypsin (Gibco, #25200-056), counted and 1.2 x 10 7 cells were plated in a Petri dish (Corning, #430167) with 10 ml of DMEM/F12 medium supplemented so that the confluence was 95-99% on the day of the experiment.
No dia seguinte, o coquetel de DNA plasmidiais foi preparado em tubos cônicos de 15 mL pela adição dos seguintes componentes: em um primeiro tubo, foram adicionados 6,92 ug do plasmídeo psPAX2 (Addgene, #12260), 3,46 ug do plasmídeo pMD2.G (Addgene, #12259), 9,62 ug do plasmídeo de transferência compreendendo um dos polinucleotídeos da presente invenção compreendendo as SEQ ID Nos: 34, 36 ou 38, e 35 pL de P3000 para um volume final de 1 ,5 m L de Opti- MEM (Gibco, 31985070) (foram utilizados vetores de segunda geração). Em outro tubo, foram misturados 41 pL de lipofectamina 3000 (Invitrogen, L3000075) e 1.459 pL de Opti-MEM. As duas soluções foram combinadas e incubadas em temperatura ambiente por 15 minutos. Passado o tempo de incubação, todo o sobrenadante foi descartado, foram adicionados novos 5 ml_ de DMEM/F12 suplementado, adicionou- se a mistura de transfecção preparada gota a gota sobre as células HEK-293T e a placa foi incubada em estufa por 6 horas. The next day, the plasmid DNA cocktail was prepared in 15 mL conical tubes by adding the following components: in a first tube, 6.92 ug of the plasmid psPAX2 (Addgene, #12260), 3.46 ug of the pMD2.G plasmid (Addgene, #12259), 9.62 μg of the transfer plasmid comprising one of the polynucleotides of the present invention comprising SEQ ID Nos: 34, 36 or 38, and 35 μL of P3000 to a final volume of 1.5 mL of Opti-MEM (Gibco, 31985070) (second generation vectors were used). In another tube, 41 μL of Lipofectamine 3000 (Invitrogen, L3000075) and 1,459 μL of Opti-MEM were mixed. The two solutions were combined and incubated at room temperature for 15 minutes. After the incubation time, all the supernatant was discarded, new 5 ml of supplemented DMEM/F12 were added, the prepared transfection mixture was added drop by drop onto the HEK-293T cells and the plate was incubated in an oven for 6 hours.
Após esta incubação, o meio foi descartado e foram adicionados 10 mL de meio Opti-MEM suplementado com 5% SFBi e 1 % de penicilina/estreptomicina, e a placa foi novamente incubada em estufa por 48 horas. Após este período, foi feita a coleta do sobrenadante contendo as partículas virais, que foi centrifugado a 300 x g por 5 minutos a 4 °C e filtrado em filtro PES (Microlab Scientific, #S33PES045S) com poros de 0,45 pm para remoção de partículas e debris celulares do sobrenadante contendo os lentivírus. O sobrenadante virai foi aliquotado e armazenado a -80 °C até o momento da transdução dos linfócitos T ou das células NK. After this incubation, the medium was discarded and 10 mL of Opti-MEM medium supplemented with 5% SFBi and 1% penicillin/streptomycin were added, and the plate was again incubated in an oven for 48 hours. After this period, the supernatant containing the viral particles was collected, centrifuged at 300 x g for 5 minutes at 4 °C and filtered through a PES filter (Microlab Scientific, #S33PES045S) with 0.45 μm pores to remove particles and cellular debris from the supernatant containing the lentiviruses. The viral supernatant was aliquoted and stored at -80 °C until transduction of T lymphocytes or NK cells.
EXEMPLO 2 - COLETA, ISOLAMENTO E ATIVAÇÃO DE CÉLULAS T COLETA EXAMPLE 2 - COLLECTION, ISOLATION AND ACTIVATION OF T CELLS COLLECTION
Foram recrutados voluntários saudáveis, de ambos os sexos, nas dependências do Centro de Pesquisa Experimental do Instituto Israelita de Ensino e Pesquisa (HEP), localizado no Hospital Israelita Albert Einstein. Todos os participantes assinaram o Termo de Consentimento Livre e Esclarecido (TCLE). Healthy volunteers of both sexes were recruited from the Experimental Research Center of the Instituto Israelita de Ensino e Pesquisa (HEP), located at the Hospital Israelita Albert Einstein. All participants signed the Free and Informed Consent Form (FICF).
A coleta de sangue periférico foi realizada por punção venosa pela equipe especializada do Hospital Israelita Albert Einstein. Foram coletados 3 tubos de sangue (8,8 mL/tubo) em tubo com S-Monovette ACD-A (Sarstedt®, NC0504898), para obtenção das células mononucleares do sangue periférico (PBMC, do inglês Peripheral Blood Mononuclear Cell). Todas as amostras foram identificadas de forma alfanumérica antes de serem processadas no IIEP para preservar a identificação dos voluntários. Peripheral blood collection was performed by venipuncture by the specialized team at Hospital Israelita Albert Einstein. Three tubes of blood (8.8 mL/tube) were collected in a tube with S-Monovette ACD-A (Sarstedt®, NC0504898), to obtain peripheral blood mononuclear cells (PBMC). Peripheral Blood Mononuclear Cell). All samples were alphanumerically identified prior to processing at IIEP to preserve volunteer identification.
As células mononucleares do sangue periférico foram obtidas através do gradiente de concentração Ficoll-Paque (Cytiva, #17144003), o qual promove a separação de células mononucleadas de hemácias e do plasma. O sangue diluído a 1 :1 com PBS 1X foi adicionado a um tubo cônico contendo o Ficoll-Paque, os tubos foram centrifugados a 400 x g por 40 minutos, com aceleração de 4 e desaceleração a 0, e a interfase, que contém as PBMC, foi coletada e lavada com tampão fosfato- salino (do inglês, phosphate-buffered saline - PBS) (Gibco, #10010-031 ). Peripheral blood mononuclear cells were obtained using a Ficoll-Paque concentration gradient (Cytiva, #17144003), which separates mononuclear cells from red blood cells and plasma. Blood diluted 1:1 with 1X PBS was added to a conical tube containing Ficoll-Paque, the tubes were centrifuged at 400 x g for 40 min with acceleration set to 4 and deceleration set to 0, and the interphase containing PBMCs was collected and washed with phosphate-buffered saline (PBS) (Gibco, #10010-031).
A seleção de células T a partir das PBMC foi feita por seleção magnética utilizando o Pan T Cell Isolation Kit (Miltenyi, 130-096-535) e a coluna LD (Miltenyi, 130-042-901 ), seguindo as instruções do fabricante. A ativação das células T selecionadas foi feita com beads magnéticas anti-CD3/CD28 (Dynabeads, Gibco, 11161 D) na proporção 1 bead : 1 célula. As etapas de seleção e ativação também são comentadas abaixo. T cell selection from PBMC was performed by magnetic selection using the Pan T Cell Isolation Kit (Miltenyi, 130-096-535) and the LD column (Miltenyi, 130-042-901), following the manufacturer's instructions. Activation of the selected T cells was performed with anti-CD3/CD28 magnetic beads (Dynabeads, Gibco, 11161 D) at a ratio of 1 bead : 1 cell. The selection and activation steps are also discussed below.
A coleta de três tubos de sangue periférico (8,8 mL/tubo) rendeu, em média, 5,6 x 107 PBMC (4,57 x 107 - 6,57 x 107, N=5) e a seleção negativa para obtenção de células CD3+ rendeu, em média, 1 ,8 x 107 células (7,5 x 106 - 4,27 x 107, N=5). The collection of three tubes of peripheral blood (8.8 mL/tube) yielded, on average, 5.6 x 10 7 PBMC (4.57 x 10 7 - 6.57 x 10 7 , N=5) and negative selection to obtain CD3+ cells yielded, on average, 1.8 x 10 7 cells (7.5 x 10 6 - 4.27 x 10 7 , N=5).
IMUNOFENOTIPAGEM DE LlNFÓCITOS T IMMUNOPHENOTYPING OF T-Linphocytes
Para a imunofenotipagem, as células foram analisadas em D0, ou seja, no mesmo dia em que foi realizada a obtenção das células mononucleares e a seleção das células T. For immunophenotyping, the cells were analyzed on D0, that is, on the same day that the mononuclear cells were obtained and the T cells were selected.
Após a contagem das células, 5 x 105 células foram separadas para a imunofenotipagem por citometria de fluxo, utilizando um painel de 5 marcadores (Tabela 1 ). O restante das células foi mantido em cultura celular, em meio RPM1 1640 (Gibco, #11875-093) suplementado (5% Soro humano AB (Sigma-Aldrich, H4522), 1 % Pen/Strepto e 50 I.U da citocina hll_-2 (Gibco, #PHC0023) - sempre adicionada a fresco). As células separadas para imunofenotipagem foram lavadas duas vezes com 500 pL de FACS Buffer (PBS 1X com 1 % SFBi), centrifugadas a 300 x g por 5 minutos e ressuspendidas em 100 pL de FACS Buffer. Em seguida, as células foram marcadas com 1 pL do reagente de viabilidade LIVE/DEAD (Invitrogen™, L34966) e incubadas em temperatura ambiente por 20 minutos no escuro. Terminada a incubação, foram feitas duas lavagens com 500 pL de FACS Buffer e as células foram marcadas com o pool de anticorpos para imunofenotipagem. Na Tabela 1 está o painel de anticorpos utilizado para imunofenotipagem e a Figura 4 demonstra a estratégia de gates utilizada para análise. Para as marcações com os anticorpos, as células foram incubadas por 15 minutos em temperatura ambiente no escuro com os anticorpos de imunofenotipagem. Após a marcação, as células foram lavadas mais duas vezes com FACS Buffer e analisadas utilizando o citômetro Attune Nxt (Thermo Fisher Scientific). A análise foi feita pelo Software FlowJo v10.6.0. After cell counting, 5 x 10 5 cells were separated for immunophenotyping by flow cytometry using a panel of 5 markers (Table 1). The remaining cells were maintained in cell culture in RPM1 1640 medium (Gibco, #11875-093) supplemented with 5% Human AB serum (Sigma-Aldrich, H4522), 1% Pen/Strepto and 50 IU of cytokine hll_-2 (Gibco, #PHC0023) - always added fresh). Cells sorted for immunophenotyping were washed twice with 500 pL of FACS Buffer (PBS 1X with 1% SFBi), centrifuged at 300 xg for 5 minutes and resuspended in 100 pL of FACS Buffer. Then, the cells were labeled with 1 pL of LIVE/DEAD viability reagent (Invitrogen™, L34966) and incubated at room temperature for 20 minutes in the dark. After the incubation was finished, two washes were performed with 500 pL of FACS Buffer and the cells were labeled with the antibody pool for immunophenotyping. Table 1 shows the antibody panel used for immunophenotyping and Figure 4 demonstrates the gating strategy used for analysis. For antibody labeling, cells were incubated for 15 minutes at room temperature in the dark with immunophenotyping antibodies. After labeling, cells were washed twice more with FACS Buffer and analyzed using the Attune Nxt cytometer (Thermo Fisher Scientific). Analysis was performed using FlowJo Software v10.6.0.
TABELA 1 - PAINEL DE ANTICORPOS UTILIZADOS PARA A IMUNOFENOTIPAGEM DETABLE 1 - PANEL OF ANTIBODIES USED FOR IMMUNOPHENOTYPING OF
LINFÓCITOS T. T LYMPHOCYTES.
Para a estratégia de gating, inicialmente foram selecionados os linfócitos dentre a população total e, dentre esses, os singlets (isolados, unitários). Em seguida, células negativas para o marcador de viabilidade LIVE/DEAD (Invitrogen™) e positivas para o CD45+ foram selecionadas para compor leucócitos vivos. Dentre esses, foram selecionadas células CD3+ e CD56” para excluir células NK e linfócitos NKT. Por fim, foi verificada a proporção de linfócitos T CD4 e CD8. For the gating strategy, lymphocytes were initially selected from the total population and, among these, singlets (isolated, unitary). Then, cells negative for the viability marker LIVE/DEAD (Invitrogen™) and positive for CD45 + were selected to compose live leukocytes. Among these, CD3 + and CD56” cells were selected to exclude NK cells and NKT lymphocytes. Finally, the proportion of CD4 and CD8 T lymphocytes was verified.
A imunofenotipagem das células pré e pós-seleção revelou a média de 96,6% (91 ,5 - 99,8%, N=7) de células CD3+ após a seleção, sendo 62,3% destas células CD4+ (48,5 - 75,8%, N=7) e 32,5% CD8+ (21 ,2 - 47,9%, N=7). Immunophenotyping of pre- and post-selection cells revealed an average of 96.6% (91.5 - 99.8%, N=7) of CD3 + cells after selection, 62.3% of which were CD4 + cells (48.5 - 75.8%, N=7) and 32.5% CD8 + (21.2 - 47.9%, N=7).
SELEÇÃO SELECTION
A seleção com beads magnéticas resultou em uma população de células com mais de 95% de positividade para o marcador de células T (CD3+), considerada pura para a realização dos experimentos seguintes. Os resultados de imunofenotipagem de linfócitos T pré e pós-seleção com beads magnéticas são mostrados na Figura 5. Selection with magnetic beads resulted in a population of cells with more than 95% positivity for the T cell marker (CD3+), considered pure for the following experiments. The results of immunophenotyping of T lymphocytes pre- and post-selection with magnetic beads are shown in Figure 5.
ATIVAÇÃO ACTIVATION
As células selecionadas foram ativadas com Dynabeads CD3/CD28 para a manutenção e expansão in vitro da população de linfócitos T. Vinte e quatro horas após a adição das Dynabeads de ativação, foi feita a identificação dos marcadores de ativação CD25 (BD Pharmigen, 555432 e CD69 (BD Biosciences, #340560 por citometria (Figura 6). The selected cells were activated with CD3/CD28 Dynabeads for in vitro maintenance and expansion of the T lymphocyte population. Twenty-four hours after addition of the activation Dynabeads, the activation markers CD25 (BD Pharmigen, 555432) and CD69 (BD Biosciences, #340560) were identified by cytometry (Figure 6).
A Figura 6 mostra que, antes da ativação com Dynabeads, 94,2% das células não expressavam nenhum dos marcadores de ativação. Já 24 horas após a ativação, aproximadamente 90% das células apresentaram expressão de pelo menos um marcador de ativação, e quase 85% apresentaram expressão de ambos os marcadores. Desta maneira, conclui-se que os linfócitos foram devidamente ativados. Figure 6 shows that, before activation with Dynabeads, 94.2% of the cells did not express any of the activation markers. Already 24 hours after activation, approximately 90% of the cells showed expression of at least one activation marker, and almost 85% showed expression of both markers. Thus, it can be concluded that the lymphocytes were properly activated.
EXEMPLO 3 - TRANSDUÇÃO DE CÉLULAS T EXAMPLE 3 - T CELL TRANSDUCTION
Para a produção de células CAR-T, as células foram transduzidas com vetores lentivirais previamente produzidos (e.g., vide Exemplo 1 ). Para isso, 2,5 x 105 células T ativadas foram semeadas por poço em placas de 24 poços, em meio de cultura RPMI suplementado. Em seguida, foi adicionado 1 ml_ da suspensão de lentivírus (não-concentrada). A adição de polybrene a uma concentração de 8 pg/mL é considerada opcional. As placas foram homogeneizadas e incubadas em estufa de cultura celular a 37 °C, com 5% CO2 e umidade controlada por 16 horas. No dia seguinte, as células foram lavadas e semeadas novamente nas placas em RPMI suplementado. As células foram expandidas, mantendo-se a concentração de 2 x 105 células/mL até 1 x 106 células/mL de meio de cultura, com troca de meio a cada 2 dias (mais frequente quando necessário) e passagem para frascos maiores de acordo com a necessidade. For the production of CAR-T cells, cells were transduced with previously produced lentiviral vectors (e.g., see Example 1). For this, 2.5 x 10 5 activated T cells were seeded per well in 24-well plates in supplemented RPMI culture medium. Then, 1 ml of the lentivirus suspension (non-concentrated) was added. The addition of polybrene at a concentration of 8 pg/mL is considered optional. The plates were homogenized and incubated in a cell culture incubator at 37 °C, with 5% CO 2 and controlled humidity for 16 hours. The following day, the cells were washed and seeded again on the plates in supplemented RPMI. The cells were expanded, maintaining the concentration of 2 x 10 5 cells/mL up to 1 x 106 cells/mL of culture medium, with medium change every 2 days (more frequent when necessary) and transfer to larger bottles as needed.
Para a avaliação da eficiência de transdução, as células transduzidas foram marcadas o Kit BCMA CAR Detection (Creative BioLabs, CARD-LX001 ), que marca especificamente CARs direcionados ao antígeno BCMA. O kit foi utilizado para a detecção de todos os CAR da presente invenção. Para a avaliação da eficiência de transdução, também foi utilizado o kit CD19 CAR Detection (Miltenyi, 130-129-550), específico a CARs direcionados ao antígeno CD19 como controle. To evaluate transduction efficiency, transduced cells were labeled with the BCMA CAR Detection Kit (Creative BioLabs, CARD-LX001), which specifically labels CARs directed to the BCMA antigen. The kit was used to detect all CARs of the present invention. To evaluate transduction efficiency, the CD19 CAR Detection kit (Miltenyi, 130-129-550), specific to CARs directed to the CD19 antigen, was also used as a control.
Para a marcação, 5 x 105 células foram coletadas, tiveram as beads de ativação removidas magneticamente, e foram então lavadas, incubadas com 1 pL do reagente de viabilidade LIVE/DEAD (Invitrogen™) por 20 minutos em temperatura ambiente e no escuro. Em seguida, as células foram lavadas com uma solução de bloqueio (PBS 1X + 0,5% BSA), a fim de reduzir ligações inespecíficas. Após a lavagem, 2 pL do reagente de detecção BCMA CAR Detection (Creative BioLabs) ou CD19 CAR Detection (Miltenyi) foram adicionados e as células foram incubadas por 45 minutos à 4°C e no escuro (BCMA CAR Detection) ou 10 minutos em temperatura ambiente no escuro (CD19 CAR Detection). Após a incubação, as células foram novamente lavadas e foi feita a marcação com anticorpos anti-CD3 e anti-CD56 por 15 minutos em temperatura ambiente no escuro. No caso do kit CD19 CAR Detection, foi adicionado também um anticorpo secundário anti-biotina. Células T não- transduzidas marcadas foram utilizadas como controle negativo. For labeling, 5 × 10 5 cells were collected, had the activation beads magnetically removed, and were then washed, incubated with 1 pL of LIVE/DEAD viability reagent (Invitrogen™) for 20 minutes at room temperature in the dark. Then, the cells were washed with a blocking solution (PBS 1X + 0.5% BSA) in order to reduce nonspecific binding. After washing, 2 pL of BCMA CAR Detection (Creative BioLabs) or CD19 CAR Detection (Miltenyi) detection reagent were added and the cells were incubated for 45 minutes at 4°C in the dark (BCMA CAR Detection) or 10 minutes at room temperature in the dark (CD19 CAR Detection). After incubation, the cells were washed again and labeled with anti-CD3 and anti-CD56 antibodies for 15 minutes at room temperature in the dark. In the case of the CD19 CAR Detection kit, an anti-biotin secondary antibody was also added. Labeled non-transduced T cells were used as a negative control.
Por fim, as células foram lavadas, ressuspendidas em 500 pL de FACS Buffer e adquiridas no citômetro Attune Nxt (Thermo Fisher Scientific). A análise foi feita pelo Software FlowJo v10.6.0. A Figura 7 demonstra a estratégia de gates utilizada para análise de células T que expressam o CAR anti-BCMA. Finally, the cells were washed, resuspended in 500 pL of FACS Buffer and acquired on the Attune Nxt cytometer (Thermo Fisher Scientific). The analysis was performed using FlowJo Software v10.6.0. Figure 7 demonstrates the gating strategy used for the analysis of T cells expressing the anti-BCMA CAR.
Foram obtidas altas taxas de eficiência de transdução para todos osHigh transduction efficiency rates were obtained for all
CARs testados (Figura 8). Os CARs de segunda geração, anti-BCMA (de SEQ ID NO: 33, codificado pela SEQ ID NO: 34, respectivamente) e anti-CD19, apresentaram eficiências superiores a 80% (n=2). Já o CAR anti-BCMA da presente invenção compreendendo IL-15 (de SEQ ID NO: 35, codificado pela SEQ ID NO: 36, respectivamente) apresentou eficiências razoavelmente inferiores, mas ainda expressivas, com uma média de eficiência de transdução de 50% (n=2). CARs tested (Figure 8). The second-generation CARs, anti-BCMA (of SEQ ID NO: 33, encoded by SEQ ID NO: 34, respectively) and anti-CD19, presented efficiencies higher than 80% (n=2). The anti-BCMA CAR of the present invention comprising IL-15 (SEQ ID NO: 35, encoded by SEQ ID NO: 36, respectively) presented reasonably lower, but still expressive, efficiencies, with an average transduction efficiency of 50% (n=2).
EXEMPLO 4 - VIABILIDADE E EXPANSÃO DAS CÉLULAS T EXAMPLE 4 - T CELL VIABILITY AND EXPANSION
As células transduzidas e não transduzidas foram expandidas in vitro por 15 dias e acompanhadas em relação à taxa de expansão e viabilidade (Figura 9). Transduced and non-transduced cells were expanded in vitro for 15 days and monitored for expansion rate and viability (Figure 9).
Não houve diferenças expressivas tanto em relação à viabilidade quanto à taxa de expansão entre as células não transduzidas e transduzidas com os vetores CAR anti-BCMA da presente invenção (n=2). Todas as células foram capazes de proliferar em cultura, com taxas de expansão de 250 até 400 vezes em 15 dias, e com boa viabilidade ao final da expansão in vitro (>90%). There were no significant differences in either viability or expansion rate between non-transduced cells and cells transduced with the anti-BCMA CAR vectors of the present invention (n=2). All cells were able to proliferate in culture, with expansion rates of 250 to 400 times in 15 days, and with good viability at the end of in vitro expansion (>90%).
EXEMPLO 5 - ENSAIO DE CITOTOXICIDADE DE CÉLULAS T - CALCEÍNA AM EXAMPLE 5 - T-CELL CYTOTOXICITY ASSAY - CALCEIN AM
A calceína acetoximetil (AM) é um corante utilizado para determinar a viabilidade de células eucarióticas. Por ser permeante, a calceína passa para o interior de células vivas, onde passa a fluorescer após a hidrólise da porção acetoximetil por esterases intracelulares ativas. Células mortas não possuem esterases ativas, portanto, não fluorescem. Acetoxymethyl calcein (AM) is a dye used to determine the viability of eukaryotic cells. Because it is permeant, calcein passes into living cells, where it fluoresces after hydrolysis of the acetoxymethyl moiety by active intracellular esterases. Dead cells do not have active esterases and therefore do not fluoresce.
O ensaio de citotoxicidade por calceína AM foi feito a partir de 4 x 105 células alvo linhagens MM1.S (ATCC, CRL-2974) ou Nalm6 (ATCC, CRL-3273), que foram marcadas com 1 ml_ de calceína AM (ThermoFisher, C1430) a 25 nM por 30 minutos no escuro. Em seguida, as células foram lavadas duas vezes com 10 mL de DPBS + 5% SFBi, ressuspendidas em meio RPMI e 1 x 104 células foram plaqueadas por poço, em placa de 96 poços (Corning, #3799). A Figura 10 corresponde ao desenho experimental do ensaio de calceína. The calcein AM cytotoxicity assay was performed using 4 × 10 5 target cells, MM1.S (ATCC, CRL-2974) or Nalm6 (ATCC, CRL-3273) lines, which were labeled with 1 ml of calcein AM (ThermoFisher, C1430) at 25 nM for 30 min in the dark. Then, the cells were washed twice with 10 mL of DPBS + 5% SFBi, resuspended in RPMI medium and 1 × 10 4 cells were plated per well in a 96-well plate (Corning, #3799). Figure 10 corresponds to the experimental design of the calcein assay.
Linfócitos CAR-T ou não-transduzidos foram contados e adicionadas aos poços contendo as células alvo nas seguintes proporções: 1 :1 , 1 :2, 2:1 , 5:1 e 10:1 (células efetoras: células alvo). Células alvo sem a marcação com calceína, células alvo marcadas com calceína e células alvo mortas com solução de 0, 1 % triton (Sigma- Aldrich) foram utilizados como controles positivos e negativos do ensaio. Todas as condições foram adquiridas por citometria de fluxo em triplicate. CAR-T or non-transduced lymphocytes were counted and added to the wells containing the target cells in the following ratios: 1:1, 1:2, 2:1, 5:1 and 10:1 (effector cells: target cells). Target cells without calcein labeling, calcein-labeled target cells, and target cells killed with 0.1% triton solution (Sigma-Aldrich) were used as positive and negative controls of the assay. All conditions were acquired by flow cytometry in triplicate.
Células efetoras e células alvo foram incubadas por 2 a 4 h em estufa e, após a incubação, foi feita a leitura no citômetro de fluxo Attune (ThermoFisher), no canal referente ao fluorocromo FITC para identificação de células alvo vivas (marcadas com calceína-AM). Effector cells and target cells were incubated for 2 to 4 h in an incubator and, after incubation, readings were taken on the Attune flow cytometer (ThermoFisher), in the channel corresponding to the FITC fluorochrome to identify live target cells (labeled with calcein-AM).
Linfócitos CAR-T anti-BCMA (com e sem IL-15) compreendendo os CAR anti-BCMA de SEQ ID NO: 33 e 35 foram co-cultivados com células alvo MM1 S por duas horas e foi analisada a taxa de morte das células alvo. Os linfócitos CAR-T também foram co-cultivados com a linhagem Nalm6 a fim de verificar a especificidade da atividade dos linfócitos. Em adição, realizou-se co-cultivos entre linfócitos não- transduzidos para verificar a citotoxicidade basal dos linfócitos T contra as linhagens testadas, e entre linfócitos CAR-T anti-CD19 para verificar a citotoxicidade de linfócitos T modificados geneticamente de maneira não específica ao antígeno BCMA e específica ao antígeno CD19 (Figura 11 ). Anti-BCMA CAR-T cells (with and without IL-15) comprising the anti-BCMA CARs of SEQ ID NO: 33 and 35 were co-cultured with MM1 S target cells for two hours and the rate of target cell killing was analyzed. CAR-T cells were also co-cultured with the Nalm6 cell line to verify the specificity of the lymphocyte activity. In addition, co-cultures were performed between non-transduced lymphocytes to verify the basal cytotoxicity of T lymphocytes against the tested cell lines, and between anti-CD19 CAR-T cells to verify the cytotoxicity of genetically modified T lymphocytes in a non-BCMA-specific and CD19-specific manner (Figure 11).
A partir desses resultados, é possível observar que houve uma maior taxa de morte das células alvo MM1 .S quando estas foram co-cultivadas com linfócitos CAR-T anti-BCMA da presente invenção compreendendo os CAR anti-BCMA de SEQ ID NO: 33 e 35, quando comparada ao co-cultivo com linfócitos CAR-T anti-CD19 e linfócitos T não transduzidos (Figura 11 A). Interessantemente, os linfócitos CAR-T anti-BCMA com IL-15 (SEQ ID NO: 35) apresentaram maiores taxas de citotoxicidade em comparação com os linfócitos CAR-T anti-BCMA sem IL-15 (SEQ ID NO: 33), mesmo que esses últimos apresentassem maior porcentagem de expressão do CAR. From these results, it is possible to observe that there was a higher death rate of the MM1.S target cells when they were co-cultured with anti-BCMA CAR-T lymphocytes of the present invention comprising the anti-BCMA CARs of SEQ ID NO: 33 and 35, when compared to the co-culture with anti-CD19 CAR-T lymphocytes and non-transduced T lymphocytes (Figure 11 A). Interestingly, the anti-BCMA CAR-T lymphocytes with IL-15 (SEQ ID NO: 35) presented higher cytotoxicity rates compared to the anti-BCMA CAR-T lymphocytes without IL-15 (SEQ ID NO: 33), even though the latter presented a higher percentage of CAR expression.
Além disso, os linfócitos CAR-T anti-BCMA da presente invenção foram mais capazes de matar células da linhagem MM1.S do que células Nalm6 (Figura 11 B), mostrando a especificidade da ação citotóxica ao antígeno BCMA. A análise das diferentes proporções de células efetoras para células alvo permite observar que houve um aumento expressivo das taxas de morte obtidas entre as proporções menores (0,5:1 , 1 :1 e 2:1 ), porém esse aumento foi mais ameno quando analisadas as maiores proporções (5:1 e 10:1 ), evidenciando um potencial platô no potencial de morte das células. Furthermore, the anti-BCMA CAR-T lymphocytes of the present invention were more capable of killing MM1.S lineage cells than Nalm6 cells (Figure 11 B), showing the specificity of the cytotoxic action to the BCMA antigen. The analysis of the different proportions of effector cells to target cells allows us to observe that there was a significant increase in the death rates obtained between the smaller proportions (0.5:1, 1:1 and 2:1), however this increase was milder when analyzing the larger proportions (5:1 and 10:1), evidencing a potential plateau in the death potential of the cells.
EXEMPLO 6 - ENSAIO DE PRODUÇÃO DE CITOCINAS CAR-T EXAMPLE 6 - CAR-T CYTOKINE PRODUCTION ASSAY
O ensaio de produção de citocinas é realizado com o objetivo de identificar a produção das citocinas IFN-y e TNF-a nos linfócitos CAR-T anti-BCMA compreendendo o CAR anti-BCMA de SEQ ID NO: 33 e 35, quando estes foram estimulados. Para isso, quatro condições experimentais foram testadas: The cytokine production assay is performed with the aim of identifying the production of the cytokines IFN-y and TNF-a in the anti-BCMA CAR-T lymphocytes comprising the anti-BCMA CAR of SEQ ID NO: 33 and 35, when they were stimulated. For this, four experimental conditions were tested:
(1 ) um controle negativo na ausência de estimulação; (1) a negative control in the absence of stimulation;
(2) um controle positivo, em que os linfócitos foram estimulados com 1X do Stimulation Cocktail contendo PMA e lonomicina (Invitrogen, #00-4970-93); (2) a positive control, in which lymphocytes were stimulated with 1X Stimulation Cocktail containing PMA and lonomycin (Invitrogen, #00-4970-93);
(3) estimulação com a linhagem MM1 S (BCMA+); e (3) stimulation with the MM1 S lineage (BCMA + ); and
(4) estimulação com a linhagem celular Nalm6 (BCMA-). (4) stimulation with the Nalm6 cell line (BCMA-).
Em uma placa de 96 poços, foram adicionados, por poço, 1 ,5 x 105 linfócitos CAR-T anti-BCMA ou linfócitos T não transduzidos, 0,2 pL de Brefeldina A (Invitrogen, #00-4506-51 ) e 0,75 pL do anticorpo anti-CD107a (BV785, BD Biosciences). Nos poços de controle positivo, foi adicionado 1X do Cocktail Stimulation (Invitrogen) e nos poços de estimulação com células foram adicionadas 1 ,5 x 105 células MM1 .S ou Nalm6. As condições foram feitas em duplicata com um volume final de 200 pL por poço e a placa foi incubada em estufa por 6h. In a 96-well plate, 1.5 x 10 5 anti-BCMA CAR-T lymphocytes or non-transduced T lymphocytes, 0.2 pL of Brefeldin A (Invitrogen, #00-4506-51), and 0.75 pL of anti-CD107a antibody (BV785, BD Biosciences) were added per well. In the positive control wells, 1X of the Stimulation Cocktail (Invitrogen) was added, and in the cell stimulation wells, 1.5 x 10 5 MM1.S or Nalm6 cells were added. The conditions were performed in duplicate with a final volume of 200 pL per well and the plate was incubated in an oven for 6 h.
Após o tempo de incubação, as células foram passadas para tubos de citometria, os poços foram lavados com 300 pL de FACS Buffer e o conteúdo foi adicionado aos respectivos tubos. As células foram então marcadas para identificação do receptor CAR com o reagente CAR BCMA Detection (Creative BioLabs), lavadas e então marcadas com 1 pL de LIVE/DEAD (Invitrogen™) por 20 minutos. As células foram novamente lavadas e incubadas com 10 pL e 1 ,25 pL dos anticorpos anti-CD3 e anti-CD56, respectivamente, por 15 minutos. Em seguida, as células foram fixadas e permeabilizadas utilizando o kit Fixation/Permeabilization Kit (BD Biosciences, 554714) e foi feita a marcação intracelular das citocinas com 2,5 pL por amostra dos anticorpos anti-IFN-y (PE, BD Biosciences) e anti-TNF-a (PE-Cy7, Biolegend). A aquisição dos dados foi feita no citômetro Attune e a análise foi feita pelo Software FlowJo v10.6.0. Abaixo está o painel de anticorpos utilizado (Tabela 2) e a figura demonstrativa da estratégia de gating utilizada para a análise é Figura 12. After the incubation time, the cells were passed into cytometry tubes, the wells were washed with 300 pL of FACS Buffer and the contents were added to the respective tubes. The cells were then labeled for CAR receptor identification with the CAR BCMA Detection reagent (Creative BioLabs), washed and then labeled with 1 pL of LIVE/DEAD (Invitrogen™) for 20 minutes. The cells were washed again and incubated with 10 pL and 1.25 pL of the anti-CD3 antibodies. and anti-CD56, respectively, for 15 minutes. Then, the cells were fixed and permeabilized using the Fixation/Permeabilization Kit (BD Biosciences, 554714) and intracellular cytokine labeling was performed with 2.5 pL per sample of anti-IFN-y (PE, BD Biosciences) and anti-TNF-a (PE-Cy7, Biolegend) antibodies. Data acquisition was performed on the Attune cytometer and analysis was performed using FlowJo Software v10.6.0. Below is the antibody panel used (Table 2) and the figure demonstrating the gating strategy used for the analysis is Figure 12.
TABELA 2 - PAINEL DE ANTICORPOS UTILIZADOS PARA O ENSAIO DE PRODUÇÃO DE TABLE 2 - PANEL OF ANTIBODIES USED FOR THE PRODUCTION ASSAY OF
CITOCINAS. CYTOKINES.
Neste ensaio, observou-se um aumento da produção de ambas as citocinas analisadas, IFN-y e TNF-a, pelos linfócitos CAR-T anti-BCMA quando co- cultivados com a linhagem celular BCMA+ MM1.S. O mesmo não foi observado quando os linfócitos foram co-cultivados com a linhagem BCMA- Nalm6, reforçando a especificidade da resposta ao antígeno BCMA. Além disso, enquanto a produção da citocina TNF-a foi semelhante entre os dois linfócitos CAR-T anti-BCMA (com e sem IL-15), a produção de IFN-y foi maior por parte dos linfócitos com expressão exógena de IL-15 em relação àqueles sem produção exógena (Figura 13). In this assay, an increase in the production of both cytokines analyzed, IFN-y and TNF-a, was observed by anti-BCMA CAR-T lymphocytes when co-cultured with the BCMA+ MM1.S cell line. The same was not observed when the lymphocytes were co-cultured with the BCMA-Nalm6 cell line, reinforcing the specificity of the response to the BCMA antigen. Furthermore, while the production of the cytokine TNF-a was similar between the two anti-BCMA CAR-T lymphocytes (with and without IL-15), the production of IFN-y was greater by lymphocytes with exogenous expression of IL-15 compared to those without exogenous production (Figure 13).
EXEMPLO 7 - SEPARAÇÃO CELULAR, ISOLAMENTO E ATIVAÇÃO DE CÉLULAS "NATURAL KILLER” (NK) EXAMPLE 7 - CELL SEPARATION, ISOLATION AND ACTIVATION OF NATURAL KILLER (NK) CELLS
CÉLULAS NK DE CORDÃO UMBILICAL UMBILICAL CORD NK CELLS
As células NK foram obtidas de cordão umbilical de indivíduos saudáveis, provenientes do Banco de Sangue do Hospital Israelita Albert Einstein (HIAE). NK cells were obtained from the umbilical cord of healthy individuals, from the Blood Bank of Hospital Israelita Albert Einstein (HIAE).
Após o descongelamento da bolsa de sangue, as células mononucleares são isoladas pela técnica de gradiente de densidade (Ficoll Paque Plus, GE Healthcare), conforme descrito anteriormente. As células NK são purificadas a partir de um kit de isolamento de células NK (NK cell isolation Kit, human - Miltenyi Biotec, 130-092-657) seguindo as instruções do fabricante. After thawing the blood bag, the mononuclear cells are isolated by the density gradient technique (Ficoll Paque Plus, GE Healthcare), as described previously. NK cells are purified from an NK cell isolation kit (NK cell isolation Kit, human - Miltenyi Biotec, 130-092-657) following the manufacturer's instructions.
Posteriormente, as células são co-cultivadas com células feeders K562 modificadas geneticamente para expressarem em sua membrana IL-21 e 41 BB (células doadas pelo MD Anderson Cancer Center), irradiadas a 100 Gy, na proporção de 2 células feeder para 1 célula NK (2:1 feeder: NK). As células são cultivadas em meio contendo RPMI 1640 GLUTAMAX (Gibco, #61870-036) e meio Click’s (Fujifilm Irvine Scientific, #9195) suplementado com Soro Fetal Bovino inativado (SFBi) (Thermo Fisher Scientific), 1X penicilina/estreptomicina e 200 LI/mL de IL-2 (Thermo Fisher Scientific). Subsequently, the cells are co-cultured with K562 feeder cells genetically modified to express IL-21 and 41 BB on their membrane (cells donated by MD Anderson Cancer Center), irradiated at 100 Gy, in a ratio of 2 feeder cells to 1 NK cell (2:1 feeder: NK). The cells are cultured in medium containing RPMI 1640 GLUTAMAX (Gibco, #61870-036) and Click’s medium (Fujifilm Irvine Scientific, #9195) supplemented with inactivated Fetal Bovine Serum (SFBi) (Thermo Fisher Scientific), 1X penicillin/streptomycin and 200 IL/mL of IL-2 (Thermo Fisher Scientific).
CÉLULAS NK DE SANGUE PERIFÉRICO COLETA PERIPHERAL BLOOD NK CELLS COLLECTION
A coleta de sangue periférico foi realizada por punção venosa pela equipe especializada do Hospital Israelita Albert Einstein. Foram coletados 10 tubos de sangue (8,8 mL/tubo) em tubo com S-Monovette ACD-A (Sarstedt®), para obtenção das células mononucleares do sangue periférico (PBMC). Todas as amostras foram identificadas de forma alfanumérica antes de serem processadas para preservar a identificação dos voluntários. Peripheral blood collection was performed by venipuncture by the specialized team at Hospital Israelita Albert Einstein. Ten tubes of blood (8.8 mL/tube) were collected in a tube with S-Monovette ACD-A (Sarstedt®) to obtain peripheral blood mononuclear cells (PBMC). All samples were identified alphanumerically before being processed to preserve the identity of the volunteers.
As células mononucleares do sangue periférico foram obtidas por gradiente de concentração utilizando o reagente Ficoll®-Paque (Cytiva). Peripheral blood mononuclear cells were obtained by concentration gradient using Ficoll®-Paque reagent (Cytiva).
Para isso, o sangue periférico é diluído na proporção 1 :1 com PBS estéril, homogeneizado gentilmente por inversão de 5 a 8 vezes e adicionado ao reagente Ficoll®-Paque na proporção de 2:1 sangue diluído/Ficoll®-Paque. Posteriormente, os tubos contendo o sangue diluído/Ficoll®-Paque foram centrifugados a 650 x g por 40 minutos com aceleração de 4 e desaceleração a 0 e a interface contendo o PBMC foi coletada. Após a coleta do PBMC, este é centrifugado a 355 x g por 5 minutos para a concentração das células. Posteriormente, o concentrado celular é solubilizado em 50 m L de PBS estéril e centrifugado a 355 x g por 5 minutos. Após esta etapa, o concentrado celular foi solubilizado em 15 ml_ de meio RPMI 1640 contendo 10% de soro fetal bovino inativado (SFBi) e 1 % de solução de antibiótico (penicilina, 10.000 ll/mL; estreptomicina, 10.000 pg/mL). For this purpose, peripheral blood is diluted in a 1:1 ratio with sterile PBS, gently homogenized by inversion 5 to 8 times and added to Ficoll®-Paque reagent in a 2:1 diluted blood/Ficoll®-Paque ratio. Subsequently, the tubes containing the diluted blood/Ficoll®-Paque were centrifuged at 650 xg for 40 minutes with acceleration of 4 and deceleration at 0 and the interface containing the PBMC was collected. After PBMC collection, it is centrifuged at 355 xg for 5 minutes to concentrate the cells. Subsequently, the cell concentrate is solubilized in 50 mL of sterile PBS and centrifuged at 355 xg for 5 minutes. After this step, the cell concentrate was solubilized in 15 mL of RPMI 1640 medium containing 10% inactivated fetal bovine serum (BFi) and 1% antibiotic solution (penicillin, 10,000 μg/mL; streptomycin, 10,000 pg/mL).
SELEÇÃO SELECTION
A seleção de células NK a partir do PBMC foi realizada por seleção magnética utilizando o kit human NK Cell Isolation (Miltenyi Biotec, 130-092-657) e a coluna LS (Miltenyi Biotec, 130-042-401 ). Nesta etapa, as células de PBMC são primeiramente centrifugadas a 300 x g por 10 minutos para a obtenção do concentrado celular. Selection of NK cells from PBMC was performed by magnetic selection using the human NK Cell Isolation kit (Miltenyi Biotec, 130-092-657) and the LS column (Miltenyi Biotec, 130-042-401). At this step, PBMC cells are first centrifuged at 300 x g for 10 min to obtain the cell concentrate.
O volume dos reagentes de seleção foi calculado a partir da contagem total de células. Na primeira etapa, o concentrado celular foi ressuspendido em MACS buffer (Miltenyi Biotec, 130-091 -376), diluição 1 :20, na proporção de 40 pL a cada 1 ,0 x 107 células. Em seguida, foi adicionado o NK Cell Biotin-Antibody Cocktail na proporção de 10 pL a cada 1 ,0 x 107 células e incubado por 5 minutos em gelo. Posteriormente foi adicionado MACS buffer, diluição 1 :20, na proporção de 30 pL a cada 1 ,0 x 107 e NK Cell MicroBead Cocktail na proporção de 20 pL a cada 1 ,0 x 107 e incubado por 10 minutos em gelo. The volume of selection reagents was calculated from the total cell count. In the first step, the cell concentrate was resuspended in MACS buffer (Miltenyi Biotec, 130-091-376), dilution 1:20, at a ratio of 40 pL for every 1.0 x 107 cells. Then, NK Cell Biotin-Antibody Cocktail was added at a ratio of 10 pL for every 1.0 x 107 cells and incubated for 5 minutes on ice. Subsequently, MACS buffer, dilution 1:20, at a ratio of 30 pL for every 1.0 x 107 and NK Cell MicroBead Cocktail at a ratio of 20 pL for every 1.0 x 107 were added and incubated for 10 minutes on ice.
Após o tempo de incubação, a mistura de células e componentes do kit de seleção foram adicionadas à coluna LS (Miltenyi Biotec) previamente lavada com 3 mL de MACS buffer, diluição 1 :20, posicionada em suporte com campo magnético e o líquido que passou pela coluna, contendo as células NK, foi coletado. Nesta etapa, as células NK foram divididas em dois grupos com a mesma quantidade de células, sendo eles: “NK memory like” (ML), células NKs ativadas com as interleucinas 12, 15 e 18 e “NK”, células NK sem ativação com interleucinas. As células NK memory like foram utilizadas neste experimento por apresentarem resposta aprimorada quando estimuladas por citocinas (Romee et al., 2012) e por promoverem um aumento na resposta antitumoral (Dong et al., 2022). After the incubation period, the mixture of cells and components of the selection kit were added to the LS column (Miltenyi Biotec) previously washed with 3 mL of MACS buffer, dilution 1:20, positioned on a support with a magnetic field and the liquid that passed through the column, containing the NK cells, was collected. At this stage, the NK cells were divided into two groups with the same amount of cells, namely: “NK memory like” (ML), NK cells activated with interleukins 12, 15 and 18 and “NK”, NK cells without activation with interleukins. The memory like NK cells were used in this experiment because they presented an improved response when stimulated by cytokines (Romee et al., 2012) and by promoting an increase in the antitumor response (Dong et al., 2022).
ATIVAÇÃO ACTIVATION
Para a ativação das células ML, estas foram semeadas seguindo a proporção de 1 ,0 X 106 células/mL em placa de 24 poços (Corning, #3473), adicionando-se as interleucinas IL-12 (10 ng/mL), IL-15 (50 ng/mL) e IL-18 (50 ng/mL) e incubando-se por 16 horas. For activation of ML cells, they were seeded following the proportion of 1.0 X 106 cells/mL in a 24-well plate (Corning, #3473), adding the interleukins IL-12 (10 ng/mL), IL-15 (50 ng/mL) and IL-18 (50 ng/mL) and incubating for 16 hours.
Após o tempo de incubação, as células ML foram lavadas com PBS 1X e centrifugadas a 300 x g por 5 minutos. A segunda lavagem foi realizada utilizando meio de cultura RPMI+Click's + 10% SFB e centrifugação 300 x g por 5 minutos. After the incubation time, the ML cells were washed with PBS 1X and centrifuged at 300 x g for 5 minutes. The second wash was performed using RPMI+Click's culture medium + 10% SFB and centrifugation at 300 x g for 5 minutes.
Posteriormente à seleção das células NK ou ativação das células ML, estas foram co-cultivadas com células feeders K562 modificadas geneticamente para expressarem em sua membrana IL-21 e 41 BB (células doadas pelo MD Anderson Cancer Center), irradiadas a 100 Gy, na proporção de 2 células feeder para 1 célula NK (2:1 feederNK). As células foram cultivadas em meio contendo RPMI 1640 GLUTAMAX (Thermo Fisher Scientific) e meio Click’s (Fujifilm) suplementado com Soro Fetal Bovino inativado pelo calor (SFBi) (Thermo Fisher Scientific), penicilina e estreptomicina (P/S) e 200U/mL de IL-2 (Thermo Fisher Scientific). A etapa de separação do PBMC e seleção de NK foram consideradas o dia zero (D0) dos experimentos. After selection of NK cells or activation of ML cells, these were co-cultured with K562 feeder cells genetically modified to express IL-21 and 41 BB on their membrane (cells donated by MD Anderson Cancer Center), irradiated at 100 Gy, in the ratio of 2 feeder cells to 1 NK cell (2:1 feederNK). The cells were cultured in medium containing RPMI 1640 GLUTAMAX (Thermo Fisher Scientific) and Click’s medium (Fujifilm) supplemented with heat-inactivated Fetal Bovine Serum (SFBi) (Thermo Fisher Scientific), penicillin and streptomycin (P/S) and 200 U/mL of IL-2 (Thermo Fisher Scientific). The PBMC separation and NK selection step were considered day zero (D0) of the experiments.
A coleta de dez tubos de sangue periférico (8,8 mL/tubo) rendeu, em média, 5,90 x 107 de PBMC (6,26 x 107 - 5,55 x 107, N=2) e a seleção para obtenção de células NK rendeu, em média, 6,7 x 106 células (7,11 x 106 - 6,29 x 106, N=2). The collection of ten tubes of peripheral blood (8.8 mL/tube) yielded, on average, 5.90 x 10 7 of PBMC (6.26 x 10 7 - 5.55 x 10 7 , N=2) and the selection to obtain NK cells yielded, on average, 6.7 x 10 6 cells (7.11 x 10 6 - 6.29 x 10 6 , N=2).
IMUNOFENOTIPAGEM DAS CÉLULAS NK IMMUNOPHENOTYPING OF NK CELLS
A imunofenotipagem foi realizada utilizando 5,0 x 105 células por tubo através da técnica de citometria de fluxo, utilizando um painel com 3 marcadores (Tabela 3). Immunophenotyping was performed using 5.0 x 105 cells per tube using the flow cytometry technique, using a panel with 3 markers (Table 3).
TABELA 3 - PAINEL DE ANTICORPOS UTILIZADOS PARA IMUNOFENOTIPAGEM DE CÉLULAS NK TABLE 3 - PANEL OF ANTIBODIES USED FOR CELL IMMUNOPHENOTYPING NK
Para a preparação das amostras, as células foram lavadas duas vezes com 500 pL de FACS Buffer (PBS 1X com 1 % SFBi), centrifugadas a 300 x g por 5 minutos e ressuspendidas em 100 pL de FACS buffer. Em seguida, as células foram marcadas com 1 pL do reagente de viabilidade LIVE/DEAD (Invitrogen™) e incubadas em temperatura ambiente por 20 minutos no escuro. Terminado o tempo de incubação, foram realizadas duas lavagens com 500 pL de FACS Buffer, com centrifugação a 300 x g por 5 minutos e as células foram marcadas com o pool de anticorpos para imunofenotipagem (Tabela 3). A marcação foi realizada por 15 minutos em temperatura ambiente, ao abrigo da luz. Após o tempo, as células foram lavadas mais duas vezes com FACS Buffer e adquiridas no citômetro Attune Nxt (Thermo Fisher Scientific). A análise dos dados foi realizada no Software FlowJo v10.6.0. A estratégia de gates utilizada para a imunofenotipagem das células NK pré e pós-seleção é mostrada na Figura 14. For sample preparation, cells were washed twice with 500 pL of FACS Buffer (PBS 1X with 1% SFBi), centrifuged at 300 x g for 5 minutes, and resuspended in 100 pL of FACS buffer. Cells were then labeled with 1 pL of LIVE/DEAD viability reagent (Invitrogen™) and incubated at room temperature for 20 minutes in the dark. After the incubation time, two washes were performed with 500 pL of FACS Buffer, with centrifugation at 300 x g for 5 minutes, and cells were labeled with the antibody pool for immunophenotyping (Table 3). Labeling was performed for 15 minutes at room temperature, protected from light. After this time, cells were washed twice more with FACS Buffer and acquired on the Attune Nxt cytometer (Thermo Fisher Scientific). Data analysis was performed using FlowJo Software v10.6.0. The gating strategy used for pre- and post-selection NK cell immunophenotyping is shown in Figure 14.
A imunofenotipagem das células pré e pós seleção mostrou uma proporção de 39% de células CD3 negativa na pré-seleção e 62% após a seleção de células NK. As células CD56 positivas e CD16 negativas representaram 29,5% na pré- seleção e 31 % na pós-seleção. As células CD56 positivas e CD16 positivas representaram 43,9% na pré-seleção e 34,9% na pós-seleção. Immunophenotyping of pre- and post-selection cells showed a proportion of 39% of CD3-negative cells in pre-selection and 62% after NK cell selection. CD56-positive and CD16-negative cells represented 29.5% in pre-selection and 31% in post-selection. CD56-positive and CD16-positive cells represented 43.9% in pre-selection and 34.9% in post-selection.
EXEMPLO 8 - TRANSDUÇÃO DAS CÉLULAS NK EXAMPLE 8 - NK CELL TRANSDUCTION
As células NK e ML foram transduzidas 6 dias após a coleta das PBMC e seleção de NK (D5) com os vetores lentivirais compreendendo o polinucleotídeo de SEQ ID NO: 38, produzidos conforme descrito, por exemplo, no Exemplo 1. NK and ML cells were transduced 6 days after PBMC collection and NK selection (D5) with lentiviral vectors comprising the polynucleotide of SEQ ID NO: 38, produced as described, for example, in Example 1.
Inicialmente ao processo de transdução, a co-cultura de célula NK+feeder e ML+feeder foram separadas de acordo com o protocolo descrito anteriormente utilizando o kit human NK Cell Isolation (Miltenyi Biotec) e coluna LS (Miltenyi Biotec). Initially during the transduction process, the NK+feeder and ML+feeder cell co-cultures were separated according to the protocol described previously using the human NK Cell Isolation kit (Miltenyi Biotec) and LS column (Miltenyi Biotec).
O processo de transdução consistiu na adição de 1 m L da suspensão de vírus não concentrado em placas de 24 poços previamente tratadas com 1 pg/mL de RetroNectin (Takara, T100B) e incubados por 4 horas em estufa a 37 °C, atmosfera de 5% de CO2. Posteriormente ao tempo de incubação, foram semeadas 2,5 x 105 de células NK ou ML por poço utilizando meio RPMI 1640+Click’s suplementado, realizou-se centrifugação a 1.000 x g, por 1 hora a 32°C e adicionou-se 10% de SFBi. No dia posterior (D6), foi realizada a troca do meio de cultura e adicionou-se as células feeder na proporção de 1 :1 (feeder: NK). The transduction process consisted of adding 1 mL of the unconcentrated virus suspension to 24-well plates previously treated with 1 pg/mL of RetroNectin (Takara, T100B) and incubating for 4 hours in an oven at 37 °C, 5% CO2 atmosphere. After the incubation time, 2.5 x 10 5 of NK or ML cells were seeded per well using supplemented RPMI 1640+Click's medium, centrifugation was performed at 1,000 xg for 1 hour at 32 °C and 10% SFBi was added. The following day (D6), the culture medium was changed and feeder cells were added in a 1:1 ratio (feeder: NK).
A avaliação da eficiência de transdução ocorreu 10 dias após a coleta de PBMC (D9), utilizando-se o Kit BCMA CAR detection (Creative BioLabs). The evaluation of transduction efficiency occurred 10 days after PBMC collection (D9), using the BCMA CAR detection Kit (Creative BioLabs).
Para o processo de marcação, coletou-se 2,5 x 105 ou 5 x 105 células e estas foram lavadas duas vezes com 500 pL de FACS Buffer (PBS 1X com 1 % SFBi), centrifugadas a 300 x g por 5 minutos e ressuspendidas em 100 pL de FACS buffer. Em seguida, as células foram marcadas com 1 pL do reagente de viabilidade LIVE/DEAD (Invitrogen™) durante 20 minutos em temperatura ambiente e no escuro. Em seguida, as células foram lavadas com uma solução de bloqueio (PBS 1X + 0,5% BSA), a fim de reduzir ligações inespecíficas. Após a lavagem, adicionou-se 2 pL do reagente de detecção BCMA CAR Detection (Creative BioLabs) e as células foram incubadas por 45 minutos à 4°C e no escuro (BCMA CAR Detection). Após o tempo de incubação, as células foram novamente lavadas e realizou-se a marcação com anticorpos anti-CD3, anti-CD56, anti-CD16 e anti-CD32 por 15 minutos em temperatura ambiente no escuro. Por fim, as células foram lavadas, ressuspendidas em 500 pL de FACS Buffer e adquiridas no citômetro Attune Nxt (Thermo Fisher Scientific). A análise foi realizada no Software FlowJo v10.6.0. For the labeling process, 2.5 x 10 5 or 5 x 10 5 cells were collected and washed twice with 500 pL of FACS Buffer (PBS 1X with 1% SFBi), centrifuged at 300 xg for 5 minutes and resuspended in 100 pL of FACS buffer. Then, the cells were labeled with 1 pL of LIVE/DEAD viability reagent (Invitrogen™) for 20 minutes at room temperature and in the dark. Then, the cells were washed with a blocking solution (PBS 1X + 0.5% BSA) in order to reduce nonspecific binding. After washing, 2 pL of BCMA CAR Detection reagent (Creative BioLabs) was added and the cells were incubated for 45 minutes at 4°C and in the dark (BCMA CAR Detection). After the incubation time, the cells were washed again and labeled with anti-CD3, anti-CD56, anti-CD16 and anti-CD32 antibodies for 15 minutes at room temperature in the dark. Finally, the cells were washed, resuspended in 500 pL of FACS Buffer and acquired on the Attune Nxt cytometer (Thermo Fisher Scientific). The analysis was performed using FlowJo Software v10.6.0.
A Figura 15 exemplifica a estratégia de gates utilizada para análise das células NK que expressam o CAR anti-BCMA-IL-15 compreendendo a SEQ ID NO: 37. Figure 15 exemplifies the gating strategy used for analysis of NK cells expressing the anti-BCMA-IL-15 CAR comprising SEQ ID NO: 37.
Na Figura 16, pode ser observada a eficiência de transdução para as células NK e ML. Para as células NK foi observado uma taxa de transdução de cerca de 7%. Para as células NK memory like foi observado uma taxa de transdução de 10%. Em ambos os grupos não transduzidos (NT) não foram detectados a expressão do CAR. In Figure 16, the transduction efficiency for NK and ML cells can be observed. For NK cells, a transduction rate of approximately 7% was observed. For memory-like NK cells, a transduction rate of 10% was observed. In both non-transduced (NT) groups, CAR expression was not detected.
EXEMPLO 9 - VIABILIDADE E EXPANSÃO DAS CÉLULAS NK EXAMPLE 9 - VIABILITY AND EXPANSION OF NK CELLS
As células transduzidas e não transduzidas foram mantidas em cultura por 21 dias e acompanhadas quanto a taxa de expansão e viabilidade. Transduced and non-transduced cells were maintained in culture for 21 days and monitored for expansion rate and viability.
Foi observado que todos os grupos experimentais foram capazes de proliferar em cultura, com taxas de expansão entre 63 e 104 vezes em 21 dias, mantendo uma viabilidade média de 80%. Não foram observadas diferenças expressivas entre as células não transduzidas e transduzidas com os vetores CAR anti-BCMA da presente invenção compreendendo a SEQ ID NO: 38 em relação a viabilidade e a taxa de expansão. It was observed that all experimental groups were able to proliferate in culture, with expansion rates between 63 and 104 times in 21 days, maintaining an average viability of 80%. No significant differences were observed between non-transduced cells and cells transduced with the anti-BCMA CAR vectors of the present invention comprising SEQ ID NO: 38 in relation to viability and expansion rate.
A eficiência de transdução do CAR anti-BCMA-IL-15 compreendendo a SEQ ID NO: 37 para as células NK e NK memory like é mostrada na Figura 17. The transduction efficiency of anti-BCMA-IL-15 CAR comprising SEQ ID NO: 37 for NK and memory-like NK cells is shown in Figure 17.
EXEMPLO 10 - ENSAIO DE CITOTOXICIDADE DAS CÉLULAS NK - CALCEÍNA AMEXAMPLE 10 - NK CELL CYTOTOXICITY ASSAY - CALCEIN AM
Conforme discutido acima, a calceína acetoximetil (AM) passa para o interior de células vivas, onde fluoresce após a hidrólise da porção acetoximetil mediada por esterases intracelulares ativas. Células mortas não possuem esterases ativas, portanto, não fluorescem. As discussed above, acetoxymethyl calcein (AM) passes into living cells, where it fluoresces following hydrolysis of the acetoxymethyl moiety mediated by active intracellular esterases. Dead cells do not have active esterases and therefore do not fluoresce.
Para verificar os efeitos citotóxicos das células NK transduzidas e não transduzidas frente à uma célula-alvo para o BCMA (alvo do CAR produzido), 4,0 x 105 da célula-alvo MM1.S (ATCC) foram marcadas com 1 mL de calceína AM a 25 nM por 30 minutos no escuro. Em seguida, as células-alvo foram lavadas duas vezes utilizando-se 10 mL de DPBS + 5% SFBi, ressuspendidas em meio RPMI 1640 suplementado e plaqueadas em placa de 96 poços com a quantidade de 1 ,0 x 104 células/poço. To verify the cytotoxic effects of transduced and non-transduced NK cells against a BCMA target cell (target of the CAR produced), 4.0 x 10 5 of the MM1.S target cell (ATCC) were labeled with 1 mL of 25 nM calcein AM for 30 minutes in the dark. Then, the target cells were washed twice using 10 mL of DPBS + 5% SFBi, resuspended in supplemented RPMI 1640 medium and plated in a 96-well plate with the amount of 1.0 x 10 4 cells/well.
As células CAR-NK, CAR-NKML ou os respectivos grupos não- transduzidos, foram contados e adicionados aos poços contendo as células-alvo nas proporções de 0,5: 1 , 1 : 1 , 2: 1 , 5: 1 e 10: 1 (células efetoras : células alvo). Células-alvo sem a marcação com calceína, células-alvo marcadas com calceína e células-alvo mortas com solução de 0, 1 % triton foram utilizadas como controles positivos e negativos do ensaio. Todas as condições foram adquiridas por citometria de fluxo em triplicate. CAR-NK, CAR-NKML or the respective non-transduced groups were counted and added to the wells containing the target cells at the ratios of 0.5:1, 1:1, 2:1, 5:1 and 10:1 (effector cells:target cells). Target cells without calcein labeling, calcein-labeled target cells and target cells killed with 0.1% triton solution were used as positive and negative controls of the assay. All conditions were acquired by flow cytometry in triplicate.
Células efetoras e células-alvo foram incubadas por 4 h em estufa a 37 °C em atmosfera de CO2 e, após o tempo de incubação, foi realizado a aquisição no citômetro de fluxo Attune, no canal referente ao fluorocromo FITC para identificação de células alvo vivas (marcadas com calceína-AM). Effector cells and target cells were incubated for 4 h in an incubator at 37 °C in a CO2 atmosphere and, after the incubation time, acquisition was performed on the Attune flow cytometer, in the channel corresponding to the FITC fluorochrome to identify live target cells (labeled with calcein-AM).
Na Figura 18, podem ser observados os gráficos de taxa de morte celular para as células NK e ML transduzidas com o CAR anti-BCMA da presente invenção compreendendo a SEQ ID NO: 37, codificado pela SEQ ID NO: 38, e não transduzidas. As células NK e ML transduzidas promoveram uma taxa de morte superior as respectivas células não transduzidas (descrita nos gráficos como NT), com P<0,0001 para a comparação em ambos os grupos (comparação entre NK transduzida e NK não transduzida e ML transduzida e ML não transduzida). Em média, a diferença da taxa de morte entre os grupos transduzidos e não transduzidos foi de 30%. In Figure 18, the cell death rate graphs for NK and ML cells transduced with the anti-BCMA CAR of the present invention comprising SEQ ID NO: 37, encoded by SEQ ID NO: 38, and non-transduced can be observed. The transduced NK and ML cells promoted a higher death rate than the respective non-transduced cells (described in the graphs as NT), with P<0.0001 for the comparison in both groups (comparison between transduced NK and non-transduced NK and transduced ML and non-transduced ML). On average, the difference in the death rate between the transduced and non-transduced groups was 30%.
EXEMPLO 11 - ENSAIO DE PRODUÇÃO DE CITOCINAS DAS CÉLULAS NK EXAMPLE 11 - NK CELL CYTOKINE PRODUCTION ASSAY
O ensaio de produção de citocinas foi realizado com o objetivo de identificar a produção das citocinas IFN-y e TNF-a e a expressão do CD107a nas células CAR-NK e CAR-NKML anti-BCMA, quando estas foram estimuladas na presença da célula-alvo MM1 S. The cytokine production assay was performed with the aim of identifying the production of the cytokines IFN-y and TNF-a and the expression of CD107a in anti-BCMA CAR-NK and CAR-NKML cells, when these were stimulated in the presence of the target cell MM1 S.
O experimento foi composto de três condições experimentais: The experiment consisted of three experimental conditions:
(1 ) um controle negativo, ausência de estimulação; (2) um controle positivo, em que as células NK foram estimuladas com um coquetel de estimulação (eBioscience™ Cell Stimulation Cocktail, Invitrogen); e(1) a negative control, absence of stimulation; (2) a positive control, in which NK cells were stimulated with a stimulation cocktail (eBioscience™ Cell Stimulation Cocktail, Invitrogen); and
(3) estimulação com a linhagem MM1.S (BCMA+). (3) stimulation with the MM1.S lineage (BCMA + ).
Em uma placa de 96 poços, foram adicionados 1 ,5 x105 de células CAR- NK ou CAR-NKML anti-BCMA da presente invenção, em que o CAR compreende a SEQ ID NO: 37, ou seus respectivos grupos não transduzidos, 0,2 pL de Brefeldina A (Thermo Fisher) e 0,75 pL do anticorpo anti-CD107a (BV785, BD Biosciences). Nos poços de controle positivo, foi adicionado 1X do coquetel de estimulação (Invitrogen) e nos poços de estimulação com células-alvo foram adicionadas 1 ,5 x 105 células MM1 S por poço. As condições foram realizadas em triplicate, com um volume final de 200 pL por poço e a placa foi incubada em estufa a 37°C, com atmosfera de CO2 durante 6 horas. In a 96-well plate, 1.5 x10 5 of anti-BCMA CAR-NK or CAR-NKML cells of the present invention, in which the CAR comprises SEQ ID NO: 37, or their respective non-transduced groups, 0.2 pL of Brefeldin A (Thermo Fisher) and 0.75 pL of the anti-CD107a antibody (BV785, BD Biosciences) were added. In the positive control wells, 1X of the stimulation cocktail (Invitrogen) was added and in the stimulation wells with target cells, 1.5 x 10 5 MM1 S cells were added per well. The conditions were performed in triplicate, with a final volume of 200 pL per well and the plate was incubated in an oven at 37°C, with a CO 2 atmosphere for 6 hours.
Após o tempo de incubação, as células foram transferidas da placa de 96 poços para tubos de citometria, sendo cada poço experimental lavado com 300 pL de FACS Buffer. Após a passagem para o tubo de citometria, as células foram marcadas para identificação do receptor CAR com o reagente CAR BCMA Detection (Creative BioLabs) durante 45 minutos, lavadas e marcadas com 1 pL de LIVE/DEAD (Invitrogen™) por 20 minutos. After the incubation time, the cells were transferred from the 96-well plate to cytometry tubes, and each experimental well was washed with 300 pL of FACS Buffer. After being transferred to the cytometry tube, the cells were labeled for CAR receptor identification with the CAR BCMA Detection reagent (Creative BioLabs) for 45 minutes, washed and labeled with 1 pL of LIVE/DEAD (Invitrogen™) for 20 minutes.
Para a marcação com os anticorpos de superfície anti-CD3 e anti-CD56, foi realizada novamente uma lavagem e as células foram incubadas com 10 pL e 2,5 pL dos anticorpos, respectivamente, por 15 minutos em FACS Buffer, em temperatura ambiente. For labeling with anti-CD3 and anti-CD56 surface antibodies, a wash was performed again and the cells were incubated with 10 pL and 2.5 pL of the antibodies, respectively, for 15 minutes in FACS Buffer, at room temperature.
Em seguida, as células foram fixadas e permeabilizadas utilizando o kit Fixation/Permeabilization Kit (BD Biosciences, 554714) e foi realizada a marcação intracelular das citocinas anti-IFN-y (PE, BD Biosciences) e anti-TNF-a (PE-Cy7, Biolegend) utilizando 2,5 pL de anticorpo por amostra. A aquisição dos dados foi realizada no citômetro de fluxo Attune Nxt (Thermo Fisher Scientific) e a análise foi realizada no Software FlowJo v10.6.0. Abaixo está indicado o painel de anticorpos utilizado (Tabela 4) e a estratégia de gates utilizada para a análise (Figura 19). The cells were then fixed and permeabilized using the Fixation/Permeabilization Kit (BD Biosciences, 554714) and intracellular labeling of anti-IFN-y (PE, BD Biosciences) and anti-TNF-a (PE-Cy7, Biolegend) cytokines was performed using 2.5 pL of antibody per sample. Data acquisition was performed on the Attune Nxt flow cytometer (Thermo Fisher Scientific) and analysis was performed using FlowJo Software v10.6.0. The antibody panel is indicated below. used (Table 4) and the gating strategy used for the analysis (Figure 19).
TABELA 4 - PAINEL DE ANTICORPOS UTILIZADOS PARA O ENSAIO DE PRODUÇÃO DETABLE 4 - PANEL OF ANTIBODIES USED FOR THE PRODUCTION ASSAY OF
CITOCINAS DAS CÉLULAS NK NK CELL CYTOKINES
Neste experimento, não foram observadas diferenças significativas de produção de ambas as citocinas, IFN-y e TNF-a, pelas células NK e ML CAR-NK anti- BCMA-IL15 quando estimuladas por PMA + lonomicina ou quando co-cultivadas com a linhagem celular BCMA+MM1 S. Também não foram observadas diferenças na produção de CD107a para os grupos transduzidos e não transduzidos estimulados com o PMA + lonomicina ou co-cultivadas com a linhagem MM1 S (Figura 20). In this experiment, no significant differences were observed in the production of both cytokines, IFN-y and TNF-a, by anti-BCMA-IL15 CAR-NK NK and ML cells when stimulated by PMA + lonomycin or when co-cultured with the BCMA+MM1 S cell line. No differences were also observed in the production of CD107a for the transduced and non-transduced groups stimulated with PMA + lonomycin or co-cultured with the MM1 S cell line (Figure 20).
REFERÊNCIAS REFERENCES
Atkin, C., Richter, A., Sapey, E. What is the significance of monoclonal gammopathy of undetermined significance? Clinical Medicine. 2018;18(5):391 . Atkin, C., Richter, A., Sapey, E. What is the significance of monoclonal gammopathy of undetermined significance? ClinicalMedicine. 2018;18(5):391 .
Bachanova, V., Cooley, S., Defor, T.E., Verneris, M.R., Zhang, B., McKenna, D.H. et al. Clearance of acute myeloid leukemia by haploidentical natural killer cells is improved using IL-2 diphtheria toxin fusion protein. Blood. 2014; 123(25):3855-63. Bachanova, V., Cooley, S., Defor, T. E., Verneris, M. R., Zhang, B., McKenna, D. H. et al. Clearance of acute myeloid leukemia by haploidentical natural killer cells is improved using IL-2 diphtheria toxin fusion protein. Blood. 2014; 123(25):3855-63.
Barrett, D.M., Singh, N., Porter, D.L., Grupp, S.A., June, C.H. Chimeric Antigen Receptor Therapy for Cancer, 2014. Barrett, D. M., Singh, N., Porter, D. L., Grupp, S. A., June, C. H. Chimeric Antigen Receptor Therapy for Cancer, 2014.
Cappell, K.M., Kochenderfer, J.N. Long-term outcomes following CAR T cell therapy: what we know so far. Nature Reviews Clinical Oncology 2023 20:6. 2023; 20(6): 359-71. Cappell, K. M., Kochenderfer, J. N. Long-term outcomes following CAR T cell therapy: what we know so far. Nature Reviews Clinical Oncology 2023 20:6. 2023; 20(6): 359-71.
Cooley, S., Trachtenberg, E., Bergemann, T.L., Saeteurn, K., Klein, J., Le, C.T. et al. Donors with group B KIR haplotypes improve relapse-free survival after unrelated hematopoietic cell transplantation for acute myelogenous leukemia. Blood. 2009; 113(3)726-32. Cooley, S., Trachtenberg, E., Bergemann, T. L., Saeteurn, K., Klein, J., Le, CT et al. Donors with group B KIR haplotypes improve relapse-free survival after unrelated hematopoietic cell transplantation for acute myelogenous leukemia. Blood. 2009; 113(3)726-32.
Cooley, S., Weisdorf, D.J., Guethlein, L.A., Klein, J.P., Wang, T., Le, C.T. et al. Donor selection for natural killer cell receptor genes leads to superior survival after unrelated transplantation for acute myelogenous leukemia. Blood. 2010; 116(14):2411 -9. Cooley, S., Weisdorf, D. J., Guethlein, L. A., Klein, J. P., Wang, T., Le, C. T. et al. Donor selection for natural killer cell receptor genes leads to superior survival after unrelated transplantation for acute myelogenous leukemia. Blood. 2010; 116(14):2411 -9.
Cooley, S., Weisdorf, D.J., Guethlein, L.A., Klein, J.P., Wang, T., Marsh, S.G. et al. Donor killer cell lg-like receptor B haplotypes, recipient HLA-C1 , and HLA- C mismatch enhance the clinical benefit of unrelated transplantation for acute myelogenous leukemia. J Immunol. 2014;192(10):4592-600. Cooley, S., Weisdorf, D.J., Guethlein, L.A., Klein, J.P., Wang, T., Marsh, S.G. et al. Donor killer cell lg-like receptor B haplotypes, recipient HLA-C1 , and HLA-C mismatch enhance the clinical benefit of unrelated transplantation for acute myelogenous leukemia. J Immunol. 2014;192(10):4592-600.
Cowan, A.J., Green, D.J., Kwok, M., Lee, S., Coffey, D.G., Holmberg, L.A., et al. Diagnosis and Management of Multiple Myeloma: A Review. JAMA. 2022;327(5):464-77. Cowan, A. J., Green, D. J., Kwok, M., Lee, S., Coffey, D. G., Holmberg, L. A., et al. Diagnosis and Management of Multiple Myeloma: A Review. JAMA. 2022;327(5):464-77.
Curti, A., Ruggeri, L, D'Addio, A., Bontadini, A., Dan, E., Motta, M.R. et al. Successful transfer of alloreactive haploidentical KIR ligand-mismatched natural killer cells after infusion in elderly high risk acute myeloid leukemia patients. Blood. 2011 ; 118(12):3273-9. da Silva Ferreira, K.I., Farley Rafael Maciel, J. I., Paula de Souza Rego Pinto Carvalho, D.I., Campos de Azevedo, 1.1., Fortes Vitor, A.I., Antonio Ferreira Júnior, M.l. Clinical characterization and survival of patients with multiple myeloma in a state of northeast brazilian Caracterización clínica y supervivencia de pacientes con múltiple mieloma en un estado del noreste Brasil. Vol. 33, Revista Cubana de Hematologia, Inmunologia y Hemoterapia. 2017. Curti, A., Ruggeri, L, D'Addio, A., Bontadini, A., Dan, E., Motta, M. R. et al. Successful transfer of alloreactive haploidentical KIR ligand-mismatched natural killer cells after infusion in elderly high-risk acute myeloid leukemia patients. Blood. 2011; 118(12):3273-9. da Silva Ferreira, K.I., Farley Rafael Maciel, J.I., Paula de Souza Rego Pinto Carvalho, D.I., Campos de Azevedo, 1.1., Fortes Vitor, A.I., Antonio Ferreira Júnior, M.l. Clinical characterization and survival of patients with multiple myeloma in a state of northeastern Brazil. Clinical characterization and survival of patients with multiple myeloma in a state of northeastern Brazil. Vol. 33, Revista Cubana de Hematologia, Inmunologia y Hemoterapia. 2017.
Davies, J.O.J., Stringaris, K., Barrett, A.J., Rezvani, K. Opportunities and limitations of natural killer cells as adoptive therapy for malignant disease. Cytotherapy. 2014; 16(11 ): 1453-66. Davies, J.O.J., Stringaris, K., Barrett, A.J., Rezvani, K. Opportunities and limitations of natural killer cells as adoptive therapy for malignant disease. Cytotherapy. 2014; 16(11): 1453-66.
Dogan, A., Siegel, D., Tran, N., Fu, A., Fowler, J., Belani, R., et al. B-cell maturation antigen expression across hematologic cancers: a systematic literature review. Blood Cancer J. 2020; 10(6). Dong, H., Ham, J.D., Hu, G., et al. Memory-like NK cells armed with a neoepitope-specific CAR exhibit potent activity against NPM1 mutated acute myeloid leukemia. Proceedings Of The National Academy Of Sciences, [S.L.], v. 119, n. 25, 13 jun. 2022. Proceedings of the National Academy of Sciences. Dogan, A., Siegel, D., Tran, N., Fu, A., Fowler, J., Belani, R., et al. B-cell maturation antigen expression across hematologic cancers: a systematic literature review. Blood Cancer J. 2020; 10(6). Dong, H., Ham, J.D., Hu, G., et al. Memory-like NK cells armed with a neoepitope-specific CAR exhibit potent activity against NPM1 mutated acute myeloid leukemia. Proceedings Of The National Academy Of Sciences, [SL], v. 119, no. 25, 13 June. 2022. Proceedings of the National Academy of Sciences.
Essand, M., Loskog, A.S.I. Genetically engineered T cells for the treatment of cancer. Vol. 273, Journal of Internal Medicine. 2013. p. 166-81. Essand, M., Loskog, A.S.I. Genetically engineered T cells for the treatment of cancer. Vol. 273, Journal of Internal Medicine. 2013. p. 166-81.
Famili, F., Wiekmeijer, A.S., Staal, F.J. The development of T cells from stem cells in mice and humans. Future Sci OA. 2017;3(3). Famili, F., Wiekmeijer, A.S., Staal, F.J. The development of T cells from stem cells in mice and humans. Future Sci OA. 2017;3(3).
Feng, D., Sun, J. Overview of anti-BCMA CAR-T immunotherapy for multiple myeloma and relapsed I refractory multiple myeloma. Scand J Immunol. 2020; 92(2): 109-19. Feng, D., Sun, J. Overview of anti-BCMA CAR-T immunotherapy for multiple myeloma and relapsed I refractory multiple myeloma. Scand J Immunol. 2020; 92(2): 109-19.
Fesnak, A.D. The Challenge of Variability in Chimeric Antigen Receptor T cell Manufacturing. Regen Eng Transl Med. 2020;6(3):322-9. Fesnak, A.D. The Challenge of Variability in Chimeric Antigen Receptor T cell Manufacturing. Regen Eng Transl Med 2020;6(3):322-9.
Friedman, K.M., Garrett, T.E., Evans, J.W., Horton, H.M., Latimer, H.J., Seidel, S.L., et al. Effective Targeting of Multiple B-Cell Maturation Antigen-Expressing Hematological Malignances by Anti-B-Cell Maturation Antigen Chimeric Antigen Receptor T Cells. Hum Gene Then 2018;29(5):585-601. Friedman, K. M., Garrett, T. E., Evans, J. W., Horton, H. M., Latimer, H. J., Seidel, S. L., et al. Effective Targeting of Multiple B-Cell Maturation Antigen-Expressing Hematological Malignancies by Anti-B-Cell Maturation Antigen Chimeric Antigen Receptor T Cells. Hum Gene Then 2018;29(5):585-601.
Geller, M.A., Cooley, S., Judson, P.L., Ghebre, R., Carson, L.F., Argenta, P.A. et al. A phase II study of allogeneic natural killer cell therapy to treat patients with recurrent ovarian and breast cancer. Cytotherapy. 2011 ; 13(1 ):98-107. Geller, M. A., Cooley, S., Judson, P. L., Ghebre, R., Carson, L. F., Argenta, P. A. et al. A phase II study of allogeneic natural killer cell therapy to treat patients with recurrent ovarian and breast cancer. Cytotherapy. 2011; 13(1):98-107.
Goulmy, E. Human minor histocompatibility antigens: new concepts for marrow transplantation and adoptive immunotherapy. Immunol Rev. 1997; 157: 125- 40. Goulmy, E. Human minor histocompatibility antigens: new concepts for marrow transplantation and adoptive immunotherapy. Immunol Rev. 1997; 157: 125-40.
Hay, K.A., Turtle, C.J. Chimeric Antigen Receptor (CAR) T Cells: Lessons Learned from Targeting of CD19 in B-Cell Malignancies. Drugs. 2017;77(3):237-45. Hay, K.A., Turtle, C.J. Chimeric Antigen Receptor (CAR) T Cells: Lessons Learned from Targeting of CD19 in B-Cell Malignancies. Drugs. 2017;77(3):237-45.
Iliopoulou, E.G., Kountourakis, P., Karamouzis, M.V., Doufexis, D.,Iliopoulou, E.G., Kountourakis, P., Karamouzis, M.V., Doufexis, D.,
Ardavanis, A., Baxevanis, C.N. et al. A phase I trial of adoptive transfer of allogeneic natural killer cells in patients with advanced non-small cell lung cancer. Cancer Immunol Immunother. 2010;59(12):1781 -9. Ardavanis, A., Baxevanis, C.N. et al. A phase I trial of adoptive transfer of allogeneic natural killer cells in patients with advanced non-small cell lung cancer. Cancer Immunol Immunother. 2010;59(12):1781 -9.
Kerbauy, L.N., Ang, S., Liu, E., Banerjee, P.P., Wu, Y., Shaim, H., et al. Cord Blood NK Cells Engineered to Express a Humanized CD123-Targeted Chimeric Antigen Receptor (CAR) and IL-15 As Off-the-Shelf Therapy for Acute Myeloid Leukemia. Blood. 2017:4453. Kerbauy, L. N., Ang, S., Liu, E., Banerjee, P. P., Wu, Y., Shaim, H., et al. Cord Blood NK Cells Engineered to Express a Humanized CD123-Targeted Chimeric Antigen Receptor (CAR) and IL-15 As Off-the-Shelf Therapy for Acute Myeloid Leukemia. Blood. 2017:4453.
Kohler, M., Greil, C., Hudecek, M., Lonial, S., Raje, N., Wãsch, R., et al. Current developments in immunotherapy in the treatment of multiple myeloma. Vol. 124, Cancer. John Wiley and Sons Inc.; 2018. p. 2075-85. Kohler, M., Greil, C., Hudecek, M., Lonial, S., Raje, N., Wãsch, R., et al. Current developments in immunotherapy in the treatment of multiple myeloma. Vol. 124, Cancer. John Wiley and Sons Inc.; 2018. p. 2075-85.
Kyle, R.A., Larson, D.R., Therneau, T.M., Dispenzieri, A., Kumar, S., Cerhan, J.R., et al. Long-Term Follow-up of Monoclonal Gammopathy of Undetermined Significance. N Engl J Med. 2018; 378(3):241-9. Kyle, R. A., Larson, D. R., Therneau, T. M., Dispenzieri, A., Kumar, S., Cerhan, J. R., et al. Long-Term Follow-up of Monoclonal Gammopathy of Undetermined Significance. N Engl J Med. 2018; 378(3):241-9.
Lanier, L.L. NK cell receptors. Annu Rev Immunol. 1998; 16:359-93.Lanier, L.L. NK cell receptors. Annu Rev Immunol. 1998; 16:359-93.
Littman, D., Hexner, E. Cancer Immunotherapy with Chimeric Antigen Receptor (CAR) T Cells. Journal of Onco-Nephrology. 2017; 1 (3): 151 -5. Littman, D., Hexner, E. Cancer Immunotherapy with Chimeric Antigen Receptor (CAR) T Cells. Journal of Onco-Nephrology. 2017; 1 (3): 151 -5.
Liu, E., Tong, Y., Dotti, G., Shaim, H., Savoldo, B., Mukherjee, M., et al. Cord blood NK cells engineered to express IL-15 and a CD19-targeted CAR show long-term persistence and potent antitumor activity. Leukemia. 2018;32(2):520-31 . Liu, E., Tong, Y., Dotti, G., Shaim, H., Savoldo, B., Mukherjee, M., et al. Cord blood NK cells engineered to express IL-15 and a CD19-targeted CAR show long-term persistence and potent antitumor activity. Leukemia. 2018;32(2):520-31 .
Mariuzza, R.A., Agnihotri, P., Orban, J. The structural basis of T-cell receptor (TCR) activation: An enduring enigma. J Biol Chem. 2020; 295(4):914-25. Mariuzza, R.A., Agnihotri, P., Orban, J. The structural basis of T-cell receptor (TCR) activation: An enduring enigma. J Biol Chem. 2020; 295(4):914-25.
Martin, T., Usmani, S.Z., Berdeja, J.G., Agha, M., Cohen, A.D., Hari, P., et al. Ciltacabtagene Autoleucel, an Anti-B-cell Maturation Antigen Chimeric Antigen Receptor T-Cell Therapy, for Relapsed/Refractory Multiple Myeloma: CARTITUDE-1 2-Year Follow-Up. J Clin Oncol [Internet], 2023; 41 (6). Martin, T., Usmani, S. Z., Berdeja, J. G., Agha, M., Cohen, A. D., Hari, P., et al. Ciltacabtagene Autoleucel, an Anti-B-cell Maturation Antigen Chimeric Antigen Receptor T-Cell Therapy, for Relapsed/Refractory Multiple Myeloma: CARTITUDE-1 2-Year Follow-Up. J Clin Oncol [Internet], 2023; 41 (6).
Maude, S.L., Frey, N., Shaw, P.A., Aplenc, R., Barrett, D.M., Bunin. N.J., et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014; 371 (16): 1507-17. Maude, S. L., Frey, N., Shaw, P. A., Aplenc, R., Barrett, D. M., Bunin. N.J., et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014; 371 (16): 1507-17.
Mehta, R.S., Rezvani, K. Chimeric Antigen Receptor Expressing Natural Killer Cells for the Immunotherapy of Cancer. Front Immunol. 2018; 9:283. Mehta, RS, Rezvani, K. Chimeric Antigen Receptor Expressing Natural Killer Cells for the Immunotherapy of Cancer. Front Immunol. 2018; 9:283.
Miliotou, A.N., Papadopoulou, L.C. CAR T-cell Therapy: A New Era in Cancer Immunotherapy. Curr Pharm Biotechnol. 2018; 19(1 ):5— 18. Miliotou, A. N., Papadopoulou, L. C. CAR T-cell Therapy: A New Era in Cancer Immunotherapy. Curr Pharm Biotechnol. 2018; 19(1):5—18.
Miller, J.S., Soignier, Y., Panoskaltsis-Mortari, A., McNearney, S.A., Yun, G.H., Fautsch, S.K. et al. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood. 2005;105(8):3051 -7. Miller, J.S., Soignier, Y., Panoskaltsis-Mortari, A., McNearney, S.A., Yun, G.H., Fautsch, S.K. et al. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood. 2005;105(8):3051 -7.
Mohanty, R., Chowdhury, C.R., Arega, S. CAR T cell therapy: A new era for cancer treatment (Review). 2019;2183-95. Mohanty, R., Chowdhury, C.R., Arega, S. CAR T cell therapy: A new era for cancer treatment (Review). 2019;2183-95.
Munshi, N.C., Anderson, L.D., Shah, N., Madduri, D., Berdeja, J., Lonial, S., et al. Idecabtagene Vicleucel in Relapsed and Refractory Multiple Myeloma. N Engl J Med [Internet], 2021 . Munshi, N. C., Anderson, L. D., Shah, N., Madduri, D., Berdeja, J., Lonial, S., et al. Idecabtagene Vicleucel in Relapsed and Refractory Multiple Myeloma. N Engl J Med [Internet], 2021 .
Oevermann, L, Michaelis, S.U., Mezger, M., Lang, P., Toporski, J., Bertaina, A. et al. KIR B haplotype donors confer a reduced risk for relapse after haploidentical transplantation in children with ALL. Blood. 2014; 124(17):2744-7. Oevermann, L, Michaelis, S. U., Mezger, M., Lang, P., Toporski, J., Bertaina, A. et al. KIR B haplotype donors confer a reduced risk for relapse after haploidentical transplantation in children with ALL. Blood. 2014; 124(17):2744-7.
Padala, S.A., Barsouk, A., Barsouk, A., Rawla, P., Vakiti, A., Kolhe, R., et al. Epidemiology, Staging, and Management of Multiple Myeloma. Medical Sciences. 2021 Jan 20;9(1 ):3. Padala, S. A., Barsouk, A., Barsouk, A., Rawla, P., Vakiti, A., Kolhe, R., et al. Epidemiology, Staging, and Management of Multiple Myeloma. Medical Sciences. 2021 Jan 20;9(1):3.
Palumbo, A., Anderson, K. Multiple Myeloma. N Engl J Med. 2011 ;364(11 ): 1046-60. Palumbo, A., Anderson, K. Multiple Myeloma. N Engl J Med. 2011 ;364(11 ): 1046-60.
Pepe, C., Asano, E., Senna, T., Asano, E., Rosim, R. Impacto econômico do mieloma múltiplo no sistema privado de saúde do Brasil. Jornal Brasileiro de Economia da Saúde. 2018 Apr;10(1 ):9-14. Pepe, C., Asano, E., Senna, T., Asano, E., Rosim, R. Economic impact of multiple myeloma on the Brazilian private health system. Brazilian Journal of Health Economics. 2018 Apr;10(1 ):9-14.
Perez-Amill, L., Suííe, G., Antonana-Vildosola, A., Castella, M., Najjar, A., Bonet, J., et al. Preclinical development of a humanized chimeric antigen receptor against B cell maturation antigen for multiple myeloma. Haematologica. 2021 ; 106(1 ): 173-84. Perez-Amill, L., Suíe, G., Antonana-Vildosola, A., Castella, M., Najjar, A., Bonet, J., et al. Preclinical development of a humanized chimeric antigen receptor against B cell maturation antigen for multiple myeloma. Haematologica. 2021 ; 106(1): 173-84.
Picanco-castro, V., Pereira, C.G., Cristina, K., Malmegrim, R. EmergingPicanco-castro, V., Pereira, C.G., Cristina, K., Malmegrim, R. Emerging
CAR T cell therapies: clinical landscape and patent technological routes. Hum Vaccin Immunother [Internet], 2020; 16(6): 1424-33. CAR T cell therapies: clinical landscape and patent technological routes. Hum Vaccine Immunother [Internet], 2020; 16(6): 1424-33.
Ramos, C.A., Dotti, C. Chimeric antigen receptor (CAR)-engineered lymphocytes for cancer therapy. Vol. 11 , Expert Opinion on Biological Therapy. 2011 . p. 855-73. Ramos, C.A., Dotti, C. Chimeric antigen receptor (CAR)-engineered lymphocytes for cancer therapy. Vol. 11, Expert Opinion on Biological Therapy. 2011 . p. 855-73.
Ravindran, A., Bartley, A.C., Holton, S.J., Gonsalves, W.I., Kapoor, P., Siddiqui, M.A., et al. Prevalence, incidence and survival of smoldering multiple myeloma in the United States. Blood Cancer J. 2016;6(10): e486. Ravindran, A., Bartley, A. C., Holton, S. J., Gonsalves, W. I., Kapoor, P., Siddiqui, M. A., et al. Prevalence, incidence and survival of smoldering multiple myeloma in the United States. Blood Cancer J. 2016;6(10): e486.
Rodriguez-Otero, P., Prosper, F., Alfonso, A., Paiva, B., San Miguel, J.F. CAR T-Cells in Multiple Myeloma Are Ready for Prime Time. J Clin Med. 2020;9(11 ):1- 16. Rodriguez-Otero, P., Prosper, F., Alfonso, A., Paiva, B., San Miguel, J.F. CAR T-Cells in Multiple Myeloma Are Ready for Prime Time. J Clin Med. 2020;9(11):1- 16.
Romee, R., Schneider, S.E., Leong, J.W. et al. Cytokine activation induces human memory-like NK cells. Blood, [S.L.], v. 120, n. 24, p. 4751-4760, 6 dez. 2012. American Society of Hematology. Romee, R., Schneider, S. E., Leong, J. W. et al. Cytokine activation induces human memory-like NK cells. Blood, [S.L.], v. 120, no. 24, p. 4751-4760, 6 Dec. 2012. American Society of Hematology.
Rubnitz, J.E., Inaba, H., Ribeiro, R.C., Pounds, S., Rooney, B., Bell, T. et al. NKAML: a pilot study to determine the safety and feasibility of haploidentical natural killer cell transplantation in childhood acute myeloid leukemia. J Clin Oncol. 2010;28(6):955-9. Rubnitz, J. E., Inaba, H., Ribeiro, R. C., Pounds, S., Rooney, B., Bell, T. et al. NKAML: a pilot study to determine the safety and feasibility of haploidentical natural killer cell transplantation in childhood acute myeloid leukemia. J Clin Oncol. 2010;28(6):955-9.
Ruggeri, L., Capanni, M., Urbani, E., Perruccio, K., Shlomchik, W.D., Tosti, A., et al. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science. 2002;295(5562):2097-100. Ruggeri, L., Capanni, M., Urbani, E., Perruccio, K., Shlomchik, W. D., Tosti, A., et al. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science. 2002;295(5562):2097-100.
Sermer, D., Brentjens, R. CAR T-cell therapy: Full speed ahead. Hematol. Oncol. 2019 Jun 1 ;37(S1 ):95-100. Sermer, D., Brentjens, R. CAR T-cell therapy: Full speed ahead. Hematol. Oncol. 2019 Jun 1 ;37(S1 ):95-100.
Shah, N., Ahmed, F., Bashir, Q., Qureshi, S., Dinh, Y., Rondon, G., et al. Durable remission with salvage second autotransplants in patients with multiple myeloma. Cancer. 2012; 118(14): 3549-55. Shah, N., Ahmed, F., Bashir, Q., Qureshi, S., Dinh, Y., Rondon, G., et al. Durable remission with salvage second autotransplants in patients with multiple myeloma. Cancer. 2012; 118(14): 3549-55.
Shah, N., Martin-Antonio, B., Yang, H., Ku, S., Lee, D.A., Cooper, L.J., et al. Antigen presenting cell-mediated expansion of human umbilical cord blood yields log-scale expansion of natural killer cells with anti-myeloma activity. PLoS One. 2013;8(10): e76781. Shah, N., Martin-Antonio, B., Yang, H., Ku, S., Lee, D.A., Cooper, L.J., et al. Antigen presenting cell-mediated expansion of human umbilical cord blood yields log-scale expansion of natural killer cells with anti-myeloma activity. PLoS One. 2013;8(10): e76781.
Shah, N., Chari, A., Scott, E., Mezzi, K., llsmani, S.Z. B-cell maturation antigen (BCMA) in multiple myeloma: rationale for targeting and current therapeutic approaches. Leukemia [Internet], 2020;985-1005. Shah, N., Chari, A., Scott, E., Mezzi, K., llsmani, S.Z. B-cell maturation antigen (BCMA) in multiple myeloma: rationale for targeting and current therapeutic approaches. Leukemia [Internet], 2020;985-1005.
Sotillo, E., Barrett, D.M., Black, K.L., Bagashev, A., Oldridge, D., Wu. G., et al. Convergence of Acquired Mutations and Alternative Splicing of CD19 Enables Resistance to CART-19 Immunotherapy. Cancer Discov. 2015;5(12):1282-95. Sotillo, E., Barrett, D. M., Black, K. L., Bagashev, A., Oldridge, D., Wu. G., et al. Convergence of Acquired Mutations and Alternative Splicing of CD19 Enables Resistance to CART-19 Immunotherapy. Cancer Discov. 2015;5(12):1282-95.
Till, B.G, Jensen, M.C., Wang, J., Qian, X., Gopal, A.K., Maloney, D.G., et al. CD20-specific adoptive immunotherapy for lymphoma using a chimeric antigen receptor with both CD28 and 4-1 BB domains: pilot clinical trial results. Blood. 2012; 119(17):3940-50. Till, B. G, Jensen, M. C., Wang, J., Qian, X., Gopal, A. K., Maloney, D. G., et al. CD20-specific adoptive immunotherapy for lymphoma using a chimeric antigen receptor with both CD28 and 4-1 BB domains: pilot clinical trial results. Blood. 2012; 119(17):3940-50.
Vivier, E., Ugolini, S., Blaise, D., Chabannon, C., Brossay, L. Targeting natural killer cells and natural killer T cells in cancer. Nat Rev Immunol. 2012; 12(4):239-52. Vivier, E., Ugolini, S., Blaise, D., Chabannon, C., Brossay, L. Targeting natural killer cells and natural killer T cells in cancer. Nat Rev Immunol. 2012; 12(4):239-52.
Wang, X., Rivière, I. Clinical manufacturing of CAR T cells: foundation of a promising therapy. Mol Ther Oncolytics. 2016; 3:16015. Wei, J., Han, X., Bo, J., Han, W. Target selection for CAR-T therapy. J Hematol. Oncol. 2019; 12(1 ). Wang, X., Rivière, I. Clinical manufacturing of CAR T cells: foundation of a promising therapy. Mol Ther Oncolytics. 2016; 3:16015. Wei, J., Han, X., Bo, J., Han, W. Target selection for CAR-T therapy. J Hematol. Oncol. 2019; 12(1).
Wilkins, O., Keeler, A.M., Flotte, T.R. CAR T-Cell Therapy: Progress and Prospects. Vol. 28, Human Gene Therapy Methods. Mary Ann Liebert Inc.; 2017. p. 61-6. Wilkins, O., Keeler, A. M., Flotte, T. R. CAR T-Cell Therapy: Progress and Prospects. Vol. 28, Human Gene Therapy Methods. Mary Ann Liebert Inc.; 2017. p. 61-6.
Zhang, C., Liu, J., Zhong, J.F., Zhang, X. Engineering CAR-T cells. Biomark Res. 2017;5(1 ). Zhang, C., Liu, J., Zhong, J. F., Zhang, X. Engineering CAR-T cells. Biomark Res. 2017;5(1).
Claims
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| BR1020230274722 | 2023-12-26 | ||
| BR102023027472-2A BR102023027472A2 (en) | 2023-12-26 | POLYNUCLEOTIDE, CHIMERIC ANTIGEN RECEPTOR, VECTOR, COMPOSITION, METHOD FOR PRODUCING MODIFIED EFFECTOR IMMUNE CELLS AND USE OF POLYNUCLEOTIDE |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025137751A1 true WO2025137751A1 (en) | 2025-07-03 |
Family
ID=96216261
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/BR2024/050577 Pending WO2025137751A1 (en) | 2023-12-26 | 2024-12-11 | Polynucleotide, chimeric antigen receptor, vector, composition, method for producing a modified immune effector cell and use of the polynucleotide |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2025137751A1 (en) |
Citations (9)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| BR112017001498A2 (en) * | 2014-07-24 | 2018-02-20 | Bluebird Bio, Inc. | bcma chimeric antigen receptors |
| WO2020034081A1 (en) * | 2018-08-14 | 2020-02-20 | Hrain Biotechnology Co., Ltd. | Bcma-targeting chimeric antigen receptor and uses thereof |
| BR112014024893B1 (en) * | 2012-04-11 | 2021-03-30 | The United States Of America, As Represented By The Secretary, Department Of Health And Human Services | ISOLATED OR PURIFIED NUCLEIC ACID SEQUENCE THAT CODES A CHEMICAL ANTIGEN (CAR) RECEPTOR AND ITS USE, ISOLATED OR PURIFIED CARS, VECTORS, METHODS FOR DESTROYING CANCERINE AND POLYNUCLEOTIDE CELLS |
| WO2022060806A1 (en) * | 2020-09-16 | 2022-03-24 | Obsidian Therapeutics, Inc. | Compositions and methods for expression of anti-bcma chimeric antigen receptors with small molecule-regulated il15 in t cells |
| US20230165872A1 (en) * | 2020-04-28 | 2023-06-01 | Juno Therapeutics, Inc. | Combination of bcma-directed t cell therapy and an immunomodulatory compound |
| WO2023212724A2 (en) * | 2022-04-28 | 2023-11-02 | Flagship Pioneering Innovations Vi, Llc | Compositions and methods for modulating a genome in t cells, induced pluripotent stem cells, and respiratory epithelial cells |
| WO2023230581A1 (en) * | 2022-05-25 | 2023-11-30 | Celgene Corporation | Methods of manufacturing t cell therapies |
| US20230398149A1 (en) * | 2020-11-04 | 2023-12-14 | Celgene Corporation | Car t cell therapy in patients who have had prior anti-cancer alkylator therapy |
| WO2023242434A1 (en) * | 2022-06-17 | 2023-12-21 | Gadeta B.V. | Modified immune cells |
-
2024
- 2024-12-11 WO PCT/BR2024/050577 patent/WO2025137751A1/en active Pending
Patent Citations (10)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| BR112014024893B1 (en) * | 2012-04-11 | 2021-03-30 | The United States Of America, As Represented By The Secretary, Department Of Health And Human Services | ISOLATED OR PURIFIED NUCLEIC ACID SEQUENCE THAT CODES A CHEMICAL ANTIGEN (CAR) RECEPTOR AND ITS USE, ISOLATED OR PURIFIED CARS, VECTORS, METHODS FOR DESTROYING CANCERINE AND POLYNUCLEOTIDE CELLS |
| US11827889B2 (en) * | 2012-04-11 | 2023-11-28 | The U.S.A., As Represented By The Secretary, Department Of Health And Human Services | Chimeric antigen receptors targeting B-cell maturation antigen |
| BR112017001498A2 (en) * | 2014-07-24 | 2018-02-20 | Bluebird Bio, Inc. | bcma chimeric antigen receptors |
| WO2020034081A1 (en) * | 2018-08-14 | 2020-02-20 | Hrain Biotechnology Co., Ltd. | Bcma-targeting chimeric antigen receptor and uses thereof |
| US20230165872A1 (en) * | 2020-04-28 | 2023-06-01 | Juno Therapeutics, Inc. | Combination of bcma-directed t cell therapy and an immunomodulatory compound |
| WO2022060806A1 (en) * | 2020-09-16 | 2022-03-24 | Obsidian Therapeutics, Inc. | Compositions and methods for expression of anti-bcma chimeric antigen receptors with small molecule-regulated il15 in t cells |
| US20230398149A1 (en) * | 2020-11-04 | 2023-12-14 | Celgene Corporation | Car t cell therapy in patients who have had prior anti-cancer alkylator therapy |
| WO2023212724A2 (en) * | 2022-04-28 | 2023-11-02 | Flagship Pioneering Innovations Vi, Llc | Compositions and methods for modulating a genome in t cells, induced pluripotent stem cells, and respiratory epithelial cells |
| WO2023230581A1 (en) * | 2022-05-25 | 2023-11-30 | Celgene Corporation | Methods of manufacturing t cell therapies |
| WO2023242434A1 (en) * | 2022-06-17 | 2023-12-21 | Gadeta B.V. | Modified immune cells |
Non-Patent Citations (1)
| Title |
|---|
| ZHANG BEIBEI, YANG MENGZHE, ZHANG WEIMING, LIU NING, WANG DAOGANG, JING LIANGFANG, XU NING, YANG NA, REN TAO: "Chimeric antigen receptor-based natural killer cell immunotherapy in cancer: from bench to bedside", CELL DEATH & DISEASE, vol. 15, no. 1, 1 January 2024 (2024-01-01), GB , pages 1 - 18, XP093331248, ISSN: 2041-4889, DOI: 10.1038/s41419-024-06438-7 * |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| Glienke et al. | GMP-compliant manufacturing of TRUCKs: CAR T cells targeting GD2 and releasing inducible IL-18 | |
| US11903967B2 (en) | Method of preparing T cells with increased activity | |
| KR20200099132A (en) | Process for producing a therapeutic composition of engineered cells | |
| EP3291835B1 (en) | Nkt-cell subset for in vivo persistence and therapeutic activity and ppropagation of same | |
| US20250002855A1 (en) | Methods for expanding t cells for the treatment of cancer and related malignancies | |
| US20190211292A1 (en) | Perfusion bioreactor bag assemblies | |
| US20250107998A1 (en) | Compositions and methods for treating immunological dysfunction | |
| CN111683971A (en) | Pharmaceutical recombinant receptor composition and method | |
| US20200330983A1 (en) | Closed-system cryogenic vessels | |
| US20240117309A1 (en) | Methods for expanding t cell populations | |
| US20220280564A1 (en) | Methods for expanding t cells for the treatment of cancer and related malignancies | |
| WO2025137751A1 (en) | Polynucleotide, chimeric antigen receptor, vector, composition, method for producing a modified immune effector cell and use of the polynucleotide | |
| BR102023027472A2 (en) | POLYNUCLEOTIDE, CHIMERIC ANTIGEN RECEPTOR, VECTOR, COMPOSITION, METHOD FOR PRODUCING MODIFIED EFFECTOR IMMUNE CELLS AND USE OF POLYNUCLEOTIDE | |
| CN117778316A (en) | Gamma delta T cell culture medium and method for preparing gamma delta T cells | |
| US20220233596A1 (en) | Modified NK-92 cells, and therapeutic and diagnostic uses thereof | |
| CN114045309A (en) | A general preparation method and application of chimeric antigen receptor-regulated T cells | |
| US20250101467A1 (en) | Preparation technique for universal car-t cell, and use of universal car-t cell thereof | |
| US20250345427A1 (en) | Method | |
| WO2024153124A1 (en) | Modified primary t cell and use thereof | |
| CN116410930A (en) | Preparation technology and application of universal CAR-T cell | |
| JP2022132969A (en) | Mrp3-targeting gene-modified t cells | |
| Santos | Towards GMP production of γðTCR engineered T cells | |
| Giunti | Immune Suppression in Multiple Myeloma: Strategies to Overcome NK Cell Inhibition | |
| HK1252164B (en) | Nkt-cell subset for in vivo persistence and therapeutic activity and ppropagation of same |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 24909251 Country of ref document: EP Kind code of ref document: A1 |
|
| DPE1 | Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101) |